Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35072208

ABSTRACT

The eye-antennal disc of Drosophila is composed of three cell layers: a columnar epithelium called the disc proper (DP); an overlying sheet of squamous cells called the peripodial epithelium (PE); and a strip of cuboidal cells that joins the other two cellular sheets to each other and comprises the outer margin (M) of the disc. The M cells play an important role in patterning the eye because it is here that the Hedgehog (Hh), Decapentaplegic (Dpp) and JAK/STAT pathways function to initiate pattern formation. Dpp signaling is lost from the margin of eyes absent (eya) mutant discs and, as a result, the initiation of retinal patterning is blocked. Based on these observations, Eya has been proposed to control the initiation of the morphogenetic furrow via regulation of Dpp signaling within the M. We show that the failure in pattern formation surprisingly results from M cells prematurely adopting a head epidermis fate. This switch in fate normally takes place during pupal development after the eye has been patterned. Our results suggest that the timing of cell fate decisions is essential for correct eye development.


Subject(s)
Compound Eye, Arthropod/cytology , Drosophila Proteins/metabolism , Eye Proteins/metabolism , Animals , Cell Differentiation , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye Proteins/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Janus Kinases/metabolism , Morphogenesis , Mutation , STAT Transcription Factors/metabolism
2.
BMC Genomics ; 25(1): 570, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844864

ABSTRACT

Compound eyes formation in decapod crustaceans occurs after the nauplius stage. However, the key genes and regulatory mechanisms of compound eye development during crustacean embryonic development have not yet been clarified. In this study, RNA-seq was used to investigate the gene expression profiles of Neocaridina denticulata sinensis from nauplius to zoea stage. Based on RNA-seq data analysis, the phototransduction and insect hormone biosynthesis pathways were enriched, and molting-related neuropeptides were highly expressed. There was strong cell proliferation in the embryo prior to compound eye development. The formation of the visual system and the hormonal regulation of hatching were the dominant biological events during compound eye development. The functional analysis of DEGs across all four developmental stages showed that cuticle formation, muscle growth and the establishment of immune system occurred from nauplius to zoea stage. Key genes related to eye development were discovered, including those involved in the determination and differentiation of the eye field, eye-color formation, and visual signal transduction. In conclusion, the results increase the understanding of the molecular mechanism of eye formation in crustacean embryonic stage.


Subject(s)
Compound Eye, Arthropod , Gene Expression Profiling , Animals , Compound Eye, Arthropod/metabolism , Compound Eye, Arthropod/growth & development , Transcriptome , Gene Expression Regulation, Developmental , Decapoda/genetics , Decapoda/growth & development , Eye/metabolism , Eye/embryology , Eye/growth & development
3.
Evol Dev ; 26(5): e12479, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38733133

ABSTRACT

Understanding the origin of novel morphological traits is a long-standing objective in evolutionary developmental biology. We explored the developmental genetic mechanisms that underpin the formation of a textbook example of evolutionary novelties, the cephalic horns of beetles. Previous work has implicated the gene regulatory networks associated with compound eye and ocellar development in horn formation and suggested that horns and compound eyes may influence each other's sizes. Therefore, we investigated the functional significance of genes central to visual system formation in the initiation, patterning, and size determination of head horns across three horned beetle species. We find that while the downregulation of canonical eye patterning genes reliably reduces or eliminates compound eye formation, it does not alter the position or shape of head horns yet does result in an increase in relative horn length. We discuss the implications of our results for our understanding of the genesis of cephalic horns in particular and evolutionary novelties in general.


Subject(s)
Coleoptera , Animals , Coleoptera/growth & development , Coleoptera/anatomy & histology , Coleoptera/genetics , Biological Evolution , Body Patterning , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/anatomy & histology , Gene Expression Regulation, Developmental , Eye/anatomy & histology , Eye/growth & development
4.
PLoS Biol ; 19(8): e3001367, 2021 08.
Article in English | MEDLINE | ID: mdl-34379617

ABSTRACT

Damage in the nervous system induces a stereotypical response that is mediated by glial cells. Here, we use the eye disc of Drosophila melanogaster as a model to explore the mechanisms involved in promoting glial cell response after neuronal cell death induction. We demonstrate that these cells rapidly respond to neuronal apoptosis by increasing in number and undergoing morphological changes, which will ultimately grant them phagocytic abilities. We found that this glial response is controlled by the activity of Decapentaplegic (Dpp) and Hedgehog (Hh) signalling pathways. These pathways are activated after cell death induction, and their functions are necessary to induce glial cell proliferation and migration to the eye discs. The latter of these 2 processes depend on the function of the c-Jun N-terminal kinase (JNK) pathway, which is activated by Dpp signalling. We also present evidence that a similar mechanism controls glial response upon apoptosis induction in the leg discs, suggesting that our results uncover a mechanism that might be involved in controlling glial cells response to neuronal cell death in different regions of the peripheral nervous system (PNS).


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Hedgehog Proteins/physiology , Neuroglia/physiology , Animals , Apoptosis , Cell Movement , Compound Eye, Arthropod/cytology , Drosophila melanogaster/cytology , MAP Kinase Signaling System
5.
Dev Biol ; 478: 173-182, 2021 10.
Article in English | MEDLINE | ID: mdl-34245727

ABSTRACT

A complex network of transcription factor interactions propagates across the larval eye disc to establish columns of evenly-spaced R8 precursor cells, the founding cells of Drosophila ommatidia. After the recruitment of additional photoreceptors to each ommatidium, the surrounding cells are organized into their stereotypical pattern during pupal development. These support cells - comprised of pigment and cone cells - are patterned to encapsulate the photoreceptors and separate ommatidia with an hexagonal honeycomb lattice. Since the proteins and processes essential for correct eye patterning are conserved, elucidating how these function and change during Drosophila eye patterning can substantially advance our understanding of transcription factor and signaling networks, cytoskeletal structures, adhesion complexes, and the biophysical properties of complex tissues during their morphogenesis. Our understanding of many of these aspects of Drosophila eye patterning is largely descriptive. Many important questions, especially relating to the regulation and integration of cellular events, remain.


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila/growth & development , Photoreceptor Cells, Invertebrate/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Compound Eye, Arthropod/cytology , Computer Simulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Larva/growth & development , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Invertebrate/cytology , Pupa/growth & development , Receptors, Invertebrate Peptide/metabolism , Signal Transduction
6.
Mol Biol Evol ; 38(5): 1924-1942, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33386848

ABSTRACT

Revealing the mechanisms underlying the breathtaking morphological diversity observed in nature is a major challenge in Biology. It has been established that recurrent mutations in hotspot genes cause the repeated evolution of morphological traits, such as body pigmentation or the gain and loss of structures. To date, however, it remains elusive whether hotspot genes contribute to natural variation in the size and shape of organs. As natural variation in head morphology is pervasive in Drosophila, we studied the molecular and developmental basis of differences in compound eye size and head shape in two closely related Drosophila species. We show differences in the progression of retinal differentiation between species and we applied comparative transcriptomics and chromatin accessibility data to identify the GATA transcription factor Pannier (Pnr) as central factor associated with these differences. Although the genetic manipulation of Pnr affected multiple aspects of dorsal head development, the effect of natural variation is restricted to a subset of the phenotypic space. We present data suggesting that this developmental constraint is caused by the coevolution of expression of pnr and its cofactor u-shaped (ush). We propose that natural variation in expression or function of highly connected developmental regulators with pleiotropic functions is a major driver for morphological evolution and we discuss implications on gene regulatory network evolution. In comparison to previous findings, our data strongly suggest that evolutionary hotspots are not the only contributors to the repeated evolution of eye size and head shape in Drosophila.


Subject(s)
Biological Evolution , Compound Eye, Arthropod/anatomy & histology , Drosophila/anatomy & histology , Genetic Pleiotropy , Animals , Compound Eye, Arthropod/growth & development , Drosophila/physiology , Drosophila Proteins/metabolism , Female , Gene Regulatory Networks , Head/anatomy & histology , Larva/growth & development , Male , Species Specificity , Transcription Factors/metabolism , Transcriptome
7.
Genesis ; 58(10-11): e23395, 2020 11.
Article in English | MEDLINE | ID: mdl-32990387

ABSTRACT

During organogenesis, cell proliferation is followed by the differentiation of specific cell types to form an organ. Any aberration in differentiation can result in developmental defects, which can result in a partial to a near-complete loss of an organ. We employ the Drosophila eye model to understand the genetic and molecular mechanisms involved in the process of differentiation. In a forward genetic screen, we identified, cullin-4 (cul-4), which encodes an E3 ubiquitin ligase, to play an important role in retinal differentiation. During development, cul-4 is known to be involved in protein degradation, regulation of genomic stability, and regulation of cell cycle. Previously, we have reported that cul-4 regulates cell death during eye development by downregulating Wingless (Wg)/Wnt signaling pathway. We found that loss-of-function of cul-4 results in a reduced eye phenotype, which can be due to onset of cell death. However, we found that loss-of-function of cul-4 also affects retinal development by downregulating retinal determination (RD) gene expression. Early markers of retinal differentiation are dysregulated in cul-4 loss of function conditions, indicating that cul-4 is necessary for differentiation. Furthermore, loss-of-function of cul-4 ectopically induces expression of negative regulators of eye development like Wg and Homothorax (Hth). During eye development, Wg is known to block the progression of a synchronous wave of differentiation referred to as Morphogenetic furrow (MF). In cul-4 loss-of-function background, expression of dpp-lacZ, a MF marker, is significantly downregulated. Our data suggest a new role of cul-4 in retinal differentiation. These studies may have significant bearings on our understanding of early eye development.


Subject(s)
Compound Eye, Arthropod/metabolism , Cullin Proteins/metabolism , Drosophila Proteins/metabolism , Neurogenesis , Animals , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/growth & development , Cullin Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation, Developmental , Loss of Function Mutation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Wnt Signaling Pathway
8.
Dev Genes Evol ; 229(1): 13-24, 2019 01.
Article in English | MEDLINE | ID: mdl-30612166

ABSTRACT

During development of higher animals, the Notch signalling pathway governs cell type specification by mediating appropriate gene expression responses. In the absence of signalling, Notch target genes are silenced by repressor complexes. In the model organism Drosophila melanogaster, the repressor complex includes the transcription factor Suppressor of Hairless [Su(H)] and Hairless (H) plus general co-repressors. Recent crystal structure analysis of the Drosophila Notch repressor revealed details of the Su(H)-H complex. They were confirmed by mutational analyses of either protein; however, only Su(H) mutants have been further studied in vivo. Here, we analyse three H variants predicted to affect Su(H) binding. To this end, amino acid replacements Phenylalanine 237, Leucines 245 and 247, as well as Tryptophan 258 to Alanine were introduced into the H protein. A cell-based reporter assay indicates substantial loss of Su(H) binding to the respective mutant proteins HFA, HLLAA and HWA. For in vivo analysis, UAS-lines HFA, HLLAA and HWA were generated to allow spatially restricted overexpression. In these assays, all three mutants resembled the HLD control, shown before to lack Su(H) binding, indicating a strong reduction of H activity. For example, the H variants were impaired in wing margin formation, but unexpectedly induced ectopic wing venation. Concurrent overexpression with Su(H), however, suggests that all mutant H protein isoforms are still able to bind Su(H) in vivo. We conclude that a weakening of the cohesion in the H-Su(H) repressor complex is sufficient for disrupting its in vivo functionality.


Subject(s)
Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Mutation , Receptors, Notch/metabolism , Transcription Factors/genetics , Animals , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster , Protein Binding , Transcription Factors/chemistry , Transcription Factors/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism
9.
Dev Biol ; 430(2): 374-384, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28645749

ABSTRACT

Cell cycle progression and differentiation are highly coordinated during the development of multicellular organisms. The mechanisms by which these processes are coordinated and how their coordination contributes to normal development are not fully understood. Here, we determine the developmental fate of a population of precursor cells in the developing Drosophila melanogaster retina that arrest in G2 phase of the cell cycle and investigate whether cell cycle phase-specific arrest influences the fate of these cells. We demonstrate that retinal precursor cells that arrest in G2 during larval development are selected as sensory organ precursors (SOPs) during pupal development and undergo two cell divisions to generate the four-cell interommatidial mechanosensory bristles. While G2 arrest is not required for bristle development, preventing G2 arrest results in incorrect bristle positioning in the adult eye. We conclude that G2-arrested cells provide a positional cue during development to ensure proper spacing of bristles in the eye. Our results suggest that the control of cell cycle progression refines cell fate decisions and that the relationship between these two processes is not necessarily deterministic.


Subject(s)
Compound Eye, Arthropod/cytology , Drosophila melanogaster/cytology , Epithelial Cells/cytology , G2 Phase , Mechanoreceptors/cytology , Animals , Cell Cycle Checkpoints/physiology , Cell Differentiation , Cell Division , Cell Lineage , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/ultrastructure , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Imaginal Discs/cytology , Larva , Mechanoreceptors/ultrastructure , Mechanotransduction, Cellular , Neuroglia/cytology , Photoreceptor Cells, Invertebrate/cytology , Pupa , Sensory Receptor Cells/cytology
10.
Biol Lett ; 14(7)2018 07.
Article in English | MEDLINE | ID: mdl-30045903

ABSTRACT

In insects, the subdivision of the head into a lateral region, harbouring the compound eyes (CEs), and a dorsal (medial) region, where the ocelli localize, is conserved. This organization might have been already present in the insects' euarthropodan ancestors. In Drosophila, the Wnt-1 homologue wingless (wg) plays a major role in the genetic subdivision of the head. To analyse specifically the role of wg signalling in the development of the dorsal head, we attenuated this pathway specifically in this region by genetic means. We find that loss of wg signalling transforms the dorsal/medial head into lateral head structures, including the development of ectopic CEs. Our genetic analysis further suggests that wg signalling organizes the dorsal head medial-lateral axis by controlling, at least in part, the expression domains of the transcription factors Otd and Ey/Pax6.


Subject(s)
Drosophila/genetics , Head/growth & development , Animals , Body Patterning , Compound Eye, Arthropod/growth & development , Drosophila/growth & development , Genes, Insect/genetics , Larva/genetics , Larva/growth & development , Signal Transduction/genetics , Transcription Factors/genetics
11.
Nature ; 490(7421): 547-51, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23023132

ABSTRACT

Mitochondrial respiratory function is frequently impaired in human cancers. However, the mechanisms by which mitochondrial dysfunction contributes to tumour progression remain elusive. Here we show in Drosophila imaginal epithelium that defects in mitochondrial function potently induce tumour progression of surrounding tissue in conjunction with oncogenic Ras. Our data show that Ras activation and mitochondrial dysfunction cooperatively stimulate production of reactive oxygen species, which causes activation of c-Jun amino (N)-terminal kinase (JNK) signalling. JNK cooperates with oncogenic Ras to inactivate the Hippo pathway, leading to upregulation of its targets Unpaired (an interleukin-6 homologue) and Wingless (a Wnt homologue). Mitochondrial dysfunction in Ras-activated cells further cooperates with Ras signalling in neighbouring cells with normal mitochondrial function, causing benign tumours to exhibit metastatic behaviour. Our findings provide a mechanistic basis for interclonal tumour progression driven by mitochondrial dysfunction and oncogenic Ras.


Subject(s)
Disease Progression , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/pathology , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Cell Transformation, Neoplastic , Clone Cells/metabolism , Clone Cells/pathology , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/pathology , Compound Eye, Arthropod/ultrastructure , Disease Models, Animal , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Imaginal Discs/metabolism , Imaginal Discs/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Up-Regulation , Wnt1 Protein/metabolism
12.
Dev Biol ; 415(1): 157-167, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27178668

ABSTRACT

Drosophila eye development is a complex process that involves many transcription factors (TFs) and interactions with their cofactors and targets. The TF Sine oculis (So) and its cofactor Eyes absent (Eya) are highly conserved and are both necessary and sufficient for eye development. Despite their many important roles during development, the direct targets of So are still largely unknown. Therefore the So-dependent regulatory network governing eye determination and differentiation is poorly understood. In this study, we intersected gene expression profiles of so or eya mutant eye tissue prepared from three different developmental stages and identified 1731 differentially expressed genes across the Drosophila genome. A combination of co-expression analyses and motif discovery identified a set of twelve putative direct So targets, including three known and nine novel targets. We also used our previous So ChIP-seq data to assess motif predictions for So and identified a canonical So binding motif. Finally, we performed in vivo enhancer reporter assays to test predicted enhancers from six candidate target genes and find that at least one enhancer from each gene is expressed in the developing eye disc and that their expression patterns overlap with that of So. We furthermore confirmed that the expression level of predicted direct So targets, for which antibodies are available, are reduced in so or eya post-mitotic knockout eye discs. In summary, we expand the set of putative So targets and show for the first time that the combined use of expression profiling of so with its cofactor eya is an effective method to identify novel So targets. Moreover, since So is highly conserved throughout the metazoa, our results provide the basis for future functional studies in a wide variety of organisms.


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Eye Proteins/physiology , Gene Expression Regulation, Developmental , Genome-Wide Association Study , Homeodomain Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Chromatin Immunoprecipitation , Compound Eye, Arthropod/ultrastructure , Consensus Sequence , Drosophila melanogaster/growth & development , Gene Ontology , Genetic Association Studies , Imaginal Discs/metabolism , Larva , Pupa , RNA, Messenger/genetics , Transcription, Genetic , Transcriptome
13.
Dev Biol ; 414(1): 4-20, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27105580

ABSTRACT

Much progress has been made in elucidating the molecular networks required for specifying retinal cells, including photoreceptors, but the downstream mechanisms that maintain identity and regulate differentiation remain poorly understood. Here, we report that the transcription factor Glass has a dual role in establishing a functional Drosophila eye. Utilizing conditional rescue approaches, we confirm that persistent defects in ommatidium patterning combined with cell death correlate with the overall disruption of eye morphology in glass mutants. In addition, we reveal that Glass exhibits a separable role in regulating photoreceptor differentiation. In particular, we demonstrate the apparent loss of glass mutant photoreceptors is not only due to cell death but also a failure of the surviving photoreceptors to complete differentiation. Moreover, the late reintroduction of Glass in these developmentally stalled photoreceptors is capable of restoring differentiation in the absence of correct ommatidium patterning. Mechanistically, transcription profiling at the time of differentiation reveals that Glass is necessary for the expression of many genes implicated in differentiation, i.e. rhabdomere morphogenesis, phototransduction, and synaptogenesis. Specifically, we show Glass directly regulates the expression of Pph13, which encodes a transcription factor necessary for opsin expression and rhabdomere morphogenesis. Finally, we demonstrate the ability of Glass to choreograph photoreceptor differentiation is conserved between Drosophila and Tribolium, two holometabolous insects. Altogether, our work identifies a fundamental regulatory mechanism to generate the full complement of cells required for a functional rhabdomeric visual system and provides a critical framework to investigate the basis of differentiation and maintenance of photoreceptor identity.


Subject(s)
Compound Eye, Arthropod/growth & development , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Photoreceptor Cells, Invertebrate/ultrastructure , Animals , Binding Sites , Cell Death , Cell Differentiation/physiology , Compound Eye, Arthropod/abnormalities , Compound Eye, Arthropod/ultrastructure , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Drosophila Proteins/biosynthesis , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Hot Temperature , Luminescent Proteins/analysis , Microscopy, Electron , Pupa , Recombinant Fusion Proteins/metabolism , Species Specificity , Transcription, Genetic , Tribolium/genetics , Tribolium/growth & development
14.
Apoptosis ; 22(4): 479-490, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28150056

ABSTRACT

The proto-oncogene Myc is well known for its roles in promoting cell growth, proliferation and apoptosis. However, in this study, we found from a genetic screen that Myc inhibits, rather than promotes, cell death triggered by c-Jun N-terminal kinase (JNK) signaling in Drosophila. Firstly, expression of Drosophila Myc (dMyc) suppresses, whereas loss of dMyc enhances, ectopically activated JNK signaling-induced cell death. Secondly, dMyc impedes physiologically activated JNK pathway-mediated cell death. Thirdly, loss of dMyc triggers JNK pathway activation and JNK-dependent cell death. Finally, the mammalian cMyc gene, when expressed in Drosophila, impedes activated JNK signaling-induced cell death. Thus, besides its well-studied apoptosis promoting function, Myc also antagonizes JNK-mediated cell death in Drosophila, and this function is likely conserved from fly to human.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Genes, myc , MAP Kinase Signaling System/genetics , Transcription Factors/physiology , Animals , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/embryology , Compound Eye, Arthropod/growth & development , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Female , Gene Expression Regulation, Developmental , Genes, Synthetic , Humans , Larva , Membrane Proteins/genetics , Membrane Proteins/physiology , Morphogenesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Recombinant Fusion Proteins/metabolism , Species Specificity , Thorax/cytology , Thorax/embryology , Thorax/growth & development , Transcription Factors/deficiency , Transcription Factors/genetics , Wings, Animal/cytology , Wings, Animal/embryology , Wings, Animal/growth & development
15.
J Insect Sci ; 17(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-28130460

ABSTRACT

In insects, some developmentally related traits are negatively correlated. Here, we mapped Quantitative Trait Loci (QTL) for traits of eye size and head capsule, in an intercontinental set of recombinant inbred lines (RILs) of Drosophila melanogaster Composite interval mapping identified QTL on all major chromosomes. Two negatively correlated traits (size of eyes and between-eyes distance) were influenced by one QTL that appeared to be antagonistic between the traits (QTL cytological range is 25F5-30A6), consistent with a negative genetic correlation between these traits of the head capsule. Comparisons of QTL across traits indicated a nonrandom distribution over the genome, with a considerable overlap between some QTL across traits. Developmentally-related traits were influenced by QTL in a pattern that is consistent both with 1) the sign of the genetic correlation between the traits and 2) a constraint in the micro-evolutionary differentiation in the traits.


Subject(s)
Compound Eye, Arthropod , Drosophila melanogaster/genetics , Quantitative Trait Loci , Animals , Compound Eye, Arthropod/growth & development , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/growth & development , Head/growth & development
16.
Dev Biol ; 407(2): 183-94, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26428511

ABSTRACT

Morphogen gradients play pervasive roles in development, and understanding how they are established and decoded is a major goal of contemporary developmental biology. Here we examine how a Wingless (Wg) morphogen gradient patterns the peripheral specialization of the fly eye. The outermost specialization is the pigment rim; a thick band of pigment cells that circumscribes the eye and optically insulates the sides of the retina. It results from the coalescence of pigment cells that survive the death of the outermost row of developing ommatidia. We investigate here how the Wg target genes expressed in the moribund ommatidia direct the intercellular signaling, the morphogenetic movements, and ultimately the ommatidial death. A salient feature of this process is the secondary expression of the Wg morphogen elicited in the ommatidia by the primary Wg signal. We find that neither the primary nor secondary sources of Wg alone are able to promote ommatidial death, but together they suffice to drive the apoptosis. This represents an unusual gradient read-out process in which a morphogen induces its own expression in its target cells to generate a concentration spike required to push the local cellular responses to the next threshold response.


Subject(s)
Apoptosis , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/growth & development , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Wnt1 Protein/metabolism , Animals , Apoptosis/genetics , Body Patterning , Compound Eye, Arthropod/ultrastructure , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Pupa/metabolism , RNA Interference , Retinal Cone Photoreceptor Cells/cytology , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/metabolism , Wnt1 Protein/genetics
17.
Mol Biol Evol ; 32(9): 2441-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25944915

ABSTRACT

Scoring the impact of noncoding variation on the function of cis-regulatory regions, on their chromatin state, and on the qualitative and quantitative expression levels of target genes is a fundamental problem in evolutionary genomics. A particular challenge is how to model the divergence of quantitative traits and to identify relationships between the changes across the different levels of the genome, the chromatin activity landscape, and the transcriptome. Here, we examine the use of the Ornstein-Uhlenbeck (OU) model to infer selection at the level of predicted cis-regulatory modules (CRMs), and link these with changes in transcription factor binding and chromatin activity. Using publicly available cross-species ChIP-Seq and STARR-Seq data we show how OU can be applied genome-wide to identify candidate transcription factors for which binding site and CRM turnover is correlated with changes in regulatory activity. Next, we profile open chromatin in the developing eye across three Drosophila species. We identify the recognition motifs of the chromatin remodelers, Trithorax-like and Grainyhead as mostly correlating with species-specific changes in open chromatin. In conclusion, we show in this study that CRM scores can be used as quantitative traits and that motif discovery approaches can be extended towards more complex models of divergence.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Regulatory Elements, Transcriptional , Transcription Factors/metabolism , Animals , Base Sequence , Compound Eye, Arthropod/growth & development , Conserved Sequence , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Genes, Insect , Genetic Speciation , Markov Chains , Models, Genetic , Phylogeny , Protein Binding , Sequence Analysis, DNA , Species Specificity , Transcription Factors/genetics
18.
Development ; 140(21): 4353-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24067351

ABSTRACT

The developing Drosophila ommatidium is characterized by two distinct waves of pattern formation. In the first wave, a precluster of five cells is formed by a complex cellular interaction mechanism. In the second wave, cells are systematically recruited to the cluster and directed to their fates by developmental cues presented by differentiating precluster cells. These developmental cues are mediated through the receptor tyrosine kinase (RTK) and Notch (N) signaling pathways and their combined activities are crucial in specifying cell type. The transcription factor Lozenge (Lz) is expressed exclusively in second wave cells. Here, we ectopically supply Lz to precluster cells and concomitantly supply the various RTK/N codes that specify each of three second wave cell fates. We thereby reproduce molecular markers of each of the second wave cell types in precluster cells and draw three inferences. First, we confirm that Lz provides key intrinsic information to second wave cells. We can now combine this with the RTK/N signaling to provide a cell fate specification code that entails both extrinsic and intrinsic information. Second, the reproduction of each second wave cell type in the precluster confirms the accuracy of the RTK/N signaling code. Third, RTK/N signaling and Lz need only be presented to the cells for a short period of time in order to specify their fate.


Subject(s)
Cell Differentiation/physiology , Compound Eye, Arthropod/growth & development , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/growth & development , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Compound Eye, Arthropod/cytology , DNA-Binding Proteins/pharmacology , Drosophila Proteins/pharmacology , Histological Techniques , Immunohistochemistry , Larva/growth & development , Transcription Factors/pharmacology
19.
Med Vet Entomol ; 30(4): 426-434, 2016 12.
Article in English | MEDLINE | ID: mdl-27670265

ABSTRACT

Bed bugs as pests of public health importance recently experienced a resurgence in populations throughout the U.S. and other countries. Consequently, recent research efforts have focused on improving understanding of bed bug physiology and behaviour to improve management. While few studies have investigated the visual capabilities of bed bugs, the present study focused specifically on eye morphology and spectral sensitivity. A 3-D imaging technique was used to document bed bug eye morphology from the first instar through adult and revealed morphological characteristics that differentiate the common bed bug from the tropical bed bug as well as sex-specific differences. Electrophysiological measurements were used to evaluate the spectral sensitivity of adult bed bugs. Male bed bugs were more responsive than females at some wavelengths. Electrophysiological studies provided evidence for at least one photoreceptor with a spectral sensitivity curve peak in the green (λmax 520 nm) region of the spectrum. The broadened long wavelength portion of the spectral sensitivity curve may potentially indicate another photoreceptor in the yellow-green (λmax 550 nm) portion of the spectrum or screening pigments. Understanding more about bed bug visual biology is vital for designing traps, which are an important component of integrated bed bug management.


Subject(s)
Bedbugs/anatomy & histology , Bedbugs/physiology , Compound Eye, Arthropod/anatomy & histology , Compound Eye, Arthropod/physiology , Animals , Bedbugs/growth & development , Compound Eye, Arthropod/growth & development , Female , Male , Nymph/anatomy & histology , Nymph/growth & development , Nymph/physiology , Vision, Ocular , Visual Perception
20.
Article in English | MEDLINE | ID: mdl-26358041

ABSTRACT

However complex a visual system is, the size (and growth rate) of all its components-lens, retina and nervous system-must be precisely tuned to each other for the system to be functional. As organisms grow, their eyes must be able to achieve and maintain emmetropia, a state in which photoreceptors receive sharp images of objects that are at infinity. While there has been ample research into how vertebrates coordinate eyes growth, this has never been addressed in arthropods with camera eyes, which tend to grow dramatically and typically in a step-wise manner with each molt (ecdysis). Here, we used histological and optical methods to measure how the larval eyes of Sunburst Diving Beetles (Thermonectus marmoratus, Coleoptera, Dytiscidae) grow, and how well optical and morphological parameters match, during the dramatic growth that occurs between two consecutive larval stages. We find that the eye tubes of the principal eyes of T. marmoratus grow substantially around molt, with the vitreous-like crystalline cone contributing the most to the overall growth. Lenses also reform relatively quickly, undergoing a period of dysfunction and then regaining the ability to project sharp images onto the retina around 8 h post-molt.


Subject(s)
Coleoptera/growth & development , Compound Eye, Arthropod/growth & development , Animals , Larva/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL