Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.679
Filter
1.
Elife ; 122024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842917

ABSTRACT

The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.


Subject(s)
Cadherins , Cell Cycle , Drosophila Proteins , Drosophila melanogaster , Signal Transduction , Wings, Animal , Animals , Wings, Animal/growth & development , Wings, Animal/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Cadherins/metabolism , Larva/growth & development , Larva/metabolism , Cell Proliferation , Cell Adhesion Molecules
2.
Biomolecules ; 14(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38785929

ABSTRACT

Suppressor of deltex (Su(dx)) is a Drosophila melanogaster member of the NEDD4 family of the HECT domain E3 ubiquitin ligases. Su(dx) acts as a regulator of Notch endocytic trafficking, promoting Notch lysosomal degradation and the down-regulation of both ligand-dependent and ligand-independent signalling, the latter involving trafficking through the endocytic pathway and activation of the endo/lysosomal membrane. Mutations of Su(dx) result in developmental phenotypes in the Drosophila wing that reflect increased Notch signalling, leading to gaps in the specification of the wing veins, and Su(dx) functions to provide the developmental robustness of Notch activity to environmental temperature shifts. The full developmental functions of Su(dx) are unclear; however, this is due to a lack of a clearly defined null allele. Here we report the first defined null mutation of Su(dx), generated by P-element excision, which removes the complete open reading frame. We show that the mutation is recessive-viable, with the Notch gain of function phenotypes affecting wing vein and leg development. We further uncover new roles for Su(dx) in Drosophila oogenesis, where it regulates interfollicular stalk formation, egg chamber separation and germline cyst enwrapment by the follicle stem cells. Interestingly, while the null allele exhibited a gain in Notch activity during oogenesis, the previously described Su(dx)SP allele, which carries a seven amino acid in-frame deletion, displayed a Notch loss of function phenotypes and an increase in follicle stem cell turnover. This is despite both alleles displaying similar Notch gain of function in wing development. We attribute this unexpected context-dependent outcome of Su(dx)sp being due to the partial retention of function by the intact C2 and WW domain regions of the protein. Our results extend our understanding of the developmental role of Su(dx) in the tissue renewal and homeostasis of the Drosophila ovary and illustrate the importance of examining an allelic series of mutations to fully understand developmental functions.


Subject(s)
Alleles , Drosophila Proteins , Drosophila melanogaster , Oogenesis , Receptors, Notch , Animals , Oogenesis/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Female , Wings, Animal/growth & development , Wings, Animal/metabolism , Mutation , Signal Transduction , Phenotype , Membrane Proteins
3.
Commun Biol ; 7(1): 533, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710747

ABSTRACT

Insect wing development is a fascinating and intricate process that involves the regulation of wing size through cell proliferation and apoptosis. In this study, we find that Ter94, an AAA-ATPase, is essential for proper wing size dependently on its ATPase activity. Loss of Ter94 enables the suppression of Hippo target genes. When Ter94 is depleted, it results in reduced wing size and increased apoptosis, which can be rescued by inhibiting the Hippo pathway. Biochemical experiments reveal that Ter94 reciprocally binds to Mer, a critical upstream component of the Hippo pathway, and disrupts its interaction with Ex and Kib. This disruption prevents the formation of the Ex-Mer-Kib complex, ultimately leading to the inactivation of the Hippo pathway and promoting proper wing development. Finally, we show that hVCP, the human homolog of Ter94, is able to substitute for Ter94 in modulating Drosophila wing size, underscoring their functional conservation. In conclusion, Ter94 plays a positive role in regulating wing size by interfering with the Ex-Mer-Kib complex, which results in the suppression of the Hippo pathway.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Membrane Proteins , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Suppressor Proteins , Wings, Animal , Animals , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Apoptosis , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
4.
Genes (Basel) ; 15(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38790181

ABSTRACT

Hairless (H) encodes the major antagonist in the Notch signaling pathway, which governs cellular differentiation of various tissues in Drosophila. By binding to the Notch signal transducer Suppressor of Hairless (Su(H)), H assembles repressor complexes onto Notch target genes. Using genome engineering, three new H alleles, HFA, HLLAA and HWA were generated and a phenotypic series was established by several parameters, reflecting the residual H-Su(H) binding capacity. Occasionally, homozygous HWA flies develop to adulthood. They were compared with the likewise semi-viable HNN allele affecting H-Su(H) nuclear entry. The H homozygotes were short-lived, sterile and flightless, yet showed largely normal expression of several mitochondrial genes. Typical for H mutants, both HWA and HNN homozygous alleles displayed strong defects in wing venation and mechano-sensory bristle development. Strikingly, however, HWA displayed only a loss of bristles, whereas bristle organs of HNN flies showed a complete shaft-to-socket transformation. Apparently, the impact of HWA is restricted to lateral inhibition, whereas that of HNN also affects the respective cell type specification. Notably, reduction in Su(H) gene dosage only suppressed the HNN bristle phenotype, but amplified that of HWA. We interpret these differences as to the role of H regarding Su(H) stability and availability.


Subject(s)
Alleles , Drosophila Proteins , Drosophila melanogaster , Wings, Animal , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Wings, Animal/growth & development , Wings, Animal/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Repressor Proteins/genetics , Repressor Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics
5.
BMC Biol ; 22(1): 98, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679694

ABSTRACT

BACKGROUND: The ability of animals to regenerate damaged tissue is a complex process that involves various cellular mechanisms. As animals age, they lose their regenerative abilities, making it essential to understand the underlying mechanisms that limit regenerative ability during aging. Drosophila melanogaster wing imaginal discs are epithelial structures that can regenerate after tissue injury. While significant research has focused on investigating regenerative responses during larval stages our comprehension of the regenerative potential of pupal wings and the underlying mechanisms contributing to the decline of regenerative responses remains limited. RESULTS: Here, we explore the temporal dynamics during pupal development of the proliferative response triggered by the induction of cell death, a typical regenerative response. Our results indicate that the apoptosis-induced proliferative response can continue until 34 h after puparium formation (APF), beyond this point cell death alone is not sufficient to induce a regenerative response. Under normal circumstances, cell proliferation ceases around 24 h APF. Interestingly, the failure of reinitiating the cell cycle beyond this time point is not attributed to an incapacity to activate the JNK pathway. Instead, our results suggest that the function of the ecdysone-responsive transcription factor E93 is involved in limiting the apoptosis-induced proliferative response during pupal development. CONCLUSIONS: Our study shows that apoptosis can prolong the proliferative period of cells in the wing during pupal development as late as 34 h APF, at least 10 h longer than during normal development. After this time point, the regenerative response is diminished, a process mediated in part by the ecdysone-responsive transcription factor E93.


Subject(s)
Apoptosis , Cell Proliferation , Drosophila Proteins , Drosophila melanogaster , Pupa , Regeneration , Transcription Factors , Wings, Animal , Animals , Wings, Animal/growth & development , Wings, Animal/physiology , Drosophila melanogaster/physiology , Drosophila melanogaster/growth & development , Pupa/growth & development , Pupa/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Regeneration/physiology
6.
Poult Sci ; 103(6): 103672, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38564834

ABSTRACT

The development of the avian wing pattern has been the subject of heated debate due to its special shape. The Suppressor of cytokine signaling 2 (SOCS2) gene encodes a negative regulator of growth hormone (GH) signaling and bone growth and is known to be strongly expressed in the third digit of chicken forelimbs. These observations suggest that SOCS2 might regulate the morphology of the avian wing, however, the function of SOCS2 in avian limb development remains unknown. Here, we reexamined SOCS2 expression in successive developmental stages of chicken limb development by in situ hybridization (ISH) and describe extended expression from the posterior of the stypolod to the third digit of the forelimbs. We used the RCAS avian retrovirus to overexpress SOCS2 in the developing chicken limb buds, which resulted in reduced or malformed chicken wings while hindlimbs developed normally. Transcriptome sequencing (mRNA-Seq) revealed changes in expression of genes known to be associated with growth and development in forelimbs with overexpressed SOCS2. This study highlights a pivotal role for SOCS2 during the development of the wing in the chicken and provides new insight into molecular mechanisms regulating avian limb development.


Subject(s)
Avian Proteins , Chickens , Suppressor of Cytokine Signaling Proteins , Wings, Animal , Animals , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Chick Embryo , Wings, Animal/growth & development , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens/growth & development , Chickens/genetics , Forelimb , Limb Buds/metabolism , Gene Expression Regulation, Developmental
7.
Cell Mol Life Sci ; 81(1): 195, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38653877

ABSTRACT

The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.


Subject(s)
Drosophila Proteins , Membrane Proteins , Myosin Heavy Chains , Receptors, Notch , Signal Transduction , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Receptors, Notch/metabolism , Receptors, Notch/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Wings, Animal/metabolism , Wings, Animal/growth & development , Drosophila/metabolism , Drosophila/genetics , Phenotype , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Cell Proliferation , Myosin Type II/metabolism , Myosin Type II/genetics
8.
J Exp Biol ; 226(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36995273

ABSTRACT

The impact of global warming on wild bee decline threatens the pollination services they provide. Exposure to temperatures above optimal during development is known to reduce adult body size but how it affects the development and scaling of body parts remains unclear. In bees, a reduction in body size and/or a reduction in body parts, such as the antennae, tongue and wings, and how they scale with body size (i.e. their allometry) could severely affect their fitness. To date, it remains unclear how temperature affects body size and the scaling of morphological traits in bees. To address this knowledge gap, we exposed both males and workers of Bombus terrestris to elevated temperature during development and assessed the effects on (i) the size of morphological traits and (ii) the allometry between these traits. Colonies were exposed to optimal (25°C) or stressful (33°C) temperatures. We then measured the body size, wing size, antenna and tongue length, as well as the allometry between these traits. We found that workers were smaller and the antennae of both castes were reduced at the higher temperature. However, tongue length and wing size were not affected by developmental temperature. The allometric scaling of the tongue was also affected by developmental temperature. Smaller body size and antennae could impair both individual and colony fitness, by affecting foraging efficiency and, consequently, colony development. Our results encourage further exploration of how the temperature-induced changes in morphology affect functional traits and pollination efficiency.


Subject(s)
Bees , Animals , Bees/growth & development , Organ Size , Body Size , Pollination , Wings, Animal/growth & development , Tongue/growth & development
9.
Science ; 379(6636): 1043-1049, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36893249

ABSTRACT

Little is known about the extent to which species use homologous regulatory architectures to achieve phenotypic convergence. By characterizing chromatin accessibility and gene expression in developing wing tissues, we compared the regulatory architecture of convergence between a pair of mimetic butterfly species. Although a handful of color pattern genes are known to be involved in their convergence, our data suggest that different mutational paths underlie the integration of these genes into wing pattern development. This is supported by a large fraction of accessible chromatin being exclusive to each species, including the de novo lineage-specific evolution of a modular optix enhancer. These findings may be explained by a high level of developmental drift and evolutionary contingency that occurs during the independent evolution of mimicry.


Subject(s)
Biological Evolution , Biological Mimicry , Butterflies , Chromatin Assembly and Disassembly , Wings, Animal , Animals , Biological Mimicry/genetics , Butterflies/anatomy & histology , Butterflies/genetics , Butterflies/growth & development , Pigmentation/genetics , Wings, Animal/anatomy & histology , Wings, Animal/growth & development , Gene Expression Regulation, Developmental , Enhancer Elements, Genetic
10.
F1000Res ; 12: 1428, 2023.
Article in English | MEDLINE | ID: mdl-38778811

ABSTRACT

Background: How the precise spatial regulation of genes is correlated with spatial variation in chromatin accessibilities is not yet clear. Previous studies that analysed chromatin from homogenates of whole-body parts of insects found little variation in chromatin accessibility across those parts, but single-cell studies of Drosophila brains showed extensive spatial variation in chromatin accessibility across that organ. In this work we studied the chromatin accessibility of butterfly wing tissue fated to differentiate distinct colors and patterns in pupal wings of Bicyclus anynana. Methods: We dissected small eyespot and adjacent control tissues from 3h pupae and performed ATAC-Seq to identify the chromatin accessibility differences between different sections of the wings. Results: We observed that three dissected wing regions showed unique chromatin accessibilities. Open chromatin regions specific to eyespot color patterns were highly enriched for binding motifs recognized by Suppressor of Hairless (Su(H)), Krüppel (Kr), Buttonhead (Btd) and Nubbin (Nub) transcription factors. Genes in the vicinity of the eyespot-specific open chromatin regions included those involved in wound healing and SMAD signal transduction pathways, previously proposed to be involved in eyespot development. Conclusions: We conclude that eyespot and non-eyespot tissue samples taken from the same wing have distinct patterns of chromatin accessibility, possibly driven by the eyespot-restricted expression of potential pioneer factors, such as Kr.


Subject(s)
Butterflies , Chromatin , Wings, Animal , Animals , Butterflies/genetics , Butterflies/metabolism , Chromatin/metabolism , Wings, Animal/metabolism , Wings, Animal/growth & development , Pigmentation , Insect Proteins/genetics , Insect Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
11.
Science ; 378(6617): 304-308, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36264807

ABSTRACT

Butterfly wing patterns derive from a deeply conserved developmental ground plan yet are diverse and evolve rapidly. It is poorly understood how gene regulatory architectures can accommodate both deep homology and adaptive change. To address this, we characterized the cis-regulatory evolution of the color pattern gene WntA in nymphalid butterflies. Comparative assay for transposase-accessible chromatin using sequencing (ATAC-seq) and in vivo deletions spanning 46 cis-regulatory elements across five species revealed deep homology of ground plan-determining sequences, except in monarch butterflies. Furthermore, noncoding deletions displayed both positive and negative regulatory effects that were often broad in nature. Our results provide little support for models predicting rapid enhancer turnover and suggest that deeply ancestral, multifunctional noncoding elements can underlie rapidly evolving trait systems.


Subject(s)
Body Patterning , Butterflies , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Pigmentation , Wings, Animal , Animals , Butterflies/genetics , Butterflies/growth & development , Pigmentation/genetics , Wings, Animal/anatomy & histology , Wings, Animal/growth & development , Body Patterning/genetics , Genetic Loci
12.
Science ; 378(6617): 249-250, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36264812
13.
PLoS One ; 17(8): e0269208, 2022.
Article in English | MEDLINE | ID: mdl-35969522

ABSTRACT

The Ajuba LIM protein Jub mediates regulation of Hippo signaling by cytoskeletal tension through interaction with the kinase Warts and participates in feedback regulation of junctional tension through regulation of the cytohesin Steppke. To investigate how Jub interacts with and regulates its distinct partners, we investigated the ability of Jub proteins missing different combinations of its three LIM domains to rescue jub phenotypes and to interact with α-catenin, Warts and Steppke. Multiple regions of Jub contribute to its ability to bind α-catenin and to localize to adherens junctions in Drosophila wing imaginal discs. Co-immunoprecipitation experiments in cultured cells identified a specific requirement for LIM2 for binding to Warts. However, in vivo, both LIM1 and LIM2, but not LIM3, were required for regulation of wing growth, Yorkie activity, and Warts localization. Conversely, LIM2 and LIM3, but not LIM1, were required for regulation of cell shape and Steppke localization in vivo, and for maximal Steppke binding in co-immunoprecipitation experiments. These observations identify distinct functions for the different LIM domains of Jub.


Subject(s)
Drosophila Proteins/physiology , Drosophila/metabolism , LIM Domain Proteins/physiology , Animals , Cytoskeleton/chemistry , Cytoskeleton/physiology , Drosophila/growth & development , Drosophila Proteins/analysis , Drosophila Proteins/genetics , LIM Domain Proteins/analysis , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/analysis , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/physiology , Signal Transduction , Transcription Factors/metabolism , Wings, Animal/growth & development , alpha Catenin/metabolism
14.
Biochem Biophys Res Commun ; 599: 87-92, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35176630

ABSTRACT

Fatty acid-binding proteins (FABPs) are lipid chaperones that mediate the intracellular dynamics of the hydrophobic molecules that they physically bind to. FABPs are implicated in sleep and psychiatric disorders, as well as in various cellular processes, such as cell proliferation and survival. FABP is well conserved in insects, and Drosophila has one FABP ortholog, dFabp, in its genome. Although dFabp appears to be evolutionarily conserved in some brain functions, little is known about its development and physiological function. In the present study, we investigated the function of dFabp in Drosophila development and behavior. Knockdown or overexpression of dFabp in the developing brain, wing, and eye resulted in developmental defects, such as decreased survival, altered cell proliferation, and increased apoptosis. Glia-specific knockdown of dFabp affected neuronal development, and neuronal regulation of dFabp affected glial cell proliferation. Moreover, the behavioral phenotypes (circadian rhythm and locomotor activity) of flies with regulated dFabp expression in glia and flies with regulated dFabp expression in neurons were very similar. Collectively, our results suggest that dFabp is involved in the development of various tissues and brain functions to control behavior and is a mediator of neuron-glia interactions in the Drosophila nervous system.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Fatty Acid-Binding Proteins/physiology , Animals , Behavior, Animal/physiology , Brain/embryology , Brain/growth & development , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Embryo, Nonmammalian/physiology , Female , Gene Expression Regulation, Developmental , Male , Wings, Animal/growth & development
15.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163119

ABSTRACT

Juvenile hormone and ecdysone are key regulators in the metamorphosis and development. Grocho (Gro) is a highly conserved protein required for metamorphosis and development. Brown planthopper (Nilaparvata lugens) is a major pest affecting rice production in China and many Asian countries. Although the molecular function of Gro has been investigated in holometabolous insects such as Aedes aegypti and Drosophila melanogaster, their role in the hemimetabolous insect, brown planthopper, and the relationship between NlGro/NlGro1-L and JH/ecdysone signaling pathway, remained unknown. In this study, NlGroucho (NlGro) and NlGroucho1-like (NlGro1-L) were cloned. An analysis of the predicted protein sequence showed that NlGro has highly conserved Q domain and WD40 domain, and NlGro1-L has a highly conserved WD40 domain. The expression profiles of both genes were studied by quantitative real-time PCR (qRT-PCR). Their relative expressions were high in egg, head, wing, ovary, and testis. NlGro and NlGro1-L were found to interact genetically with juvenile hormone and ecdysone signaling by hormone treatment and RNAi of JH/ecdysone signaling-related genes. Moreover, when NlGro or NlGro1-L was down-regulated alone, the survival rate was decreased, the ovarian development was delayed, and the oviposition was also affected. All defects were aggravated when NlGro and NlGro1-L were down-regulated together. This study will help to develop new pesticides on the basis of the function of NlGro and NlGro1-L, and provide new possibilities for the control of Nilaparvata lugens.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Hemiptera/growth & development , Insect Proteins/metabolism , Juvenile Hormones/pharmacology , Metamorphosis, Biological , Ovary/growth & development , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Hemiptera/drug effects , Hemiptera/genetics , Hemiptera/metabolism , Insect Proteins/genetics , Ovary/drug effects , Ovary/metabolism , Oviposition , Sequence Homology , Wings, Animal/drug effects , Wings, Animal/growth & development
16.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35169073

ABSTRACT

Butterfly eyespots are beautiful novel traits with an unknown developmental origin. Here we show that eyespots likely originated via cooption of parts of an ancestral appendage gene-regulatory network (GRN) to novel locations on the wing. Using comparative transcriptome analysis, we show that eyespots cluster most closely with antennae, relative to multiple other tissues. Furthermore, three genes essential for eyespot development, Distal-less (Dll), spalt (sal), and Antennapedia (Antp), share similar regulatory connections as those observed in the antennal GRN. CRISPR knockout of cis-regulatory elements (CREs) for Dll and sal led to the loss of eyespots, antennae, legs, and also wings, demonstrating that these CREs are highly pleiotropic. We conclude that eyespots likely reused an ancient GRN for their development, a network also previously implicated in the development of antennae, legs, and wings.


Subject(s)
Body Patterning/genetics , Gene Regulatory Networks/genetics , Pigmentation/genetics , Animals , Arthropod Antennae/growth & development , Biological Evolution , Butterflies/embryology , Butterflies/genetics , Evolution, Molecular , Extremities/growth & development , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Phenotype , Wings, Animal/growth & development
17.
Elife ; 112022 01 17.
Article in English | MEDLINE | ID: mdl-35037619

ABSTRACT

Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.


Subject(s)
Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Wings, Animal/growth & development , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Hedgehog Proteins/genetics , Larva/growth & development , Larva/metabolism
18.
Insect Biochem Mol Biol ; 141: 103718, 2022 02.
Article in English | MEDLINE | ID: mdl-34982980

ABSTRACT

Chitin is an aminopolysaccharide present in insects as a major structural component of the cuticle. However, current knowledge on the chitin biosynthetic machinery, especially its constituents and mechanism, is limited. Using three independent binding assays, including co-immunoprecipitation, split-ubiquitin membrane yeast two-hybrid assay, and pull-down assay, we demonstrate that choline transporter-like protein 2 (Ctl2) interacts with krotzkopf verkehrt (kkv) in Drosophila melanogaster. The global knockdown of Ctl2 by RNA interference (RNAi) induced lethality at the larval stage. Tissue-specific RNAi to silence Ctl2 in the tracheal system and in the epidermis of the flies resulted in lethality at the first larval instar. The knockdown of Ctl2 in wings led to shrunken wings containing accumulated fluid. Calcofluor White staining demonstrated reduced chitin content in the first longitudinal vein of Ctl2 knockdown wings. The pro-cuticle, which was thinner compared to wildtype, exhibited a reduced number of chitin laminar layers. Phylogenetic analyses revealed orthologues of Ctl2 in different insect orders with highly conserved domains. Our findings provide new insights into cuticle formation, wherein Ctl2 plays an important role as a chitin-synthase interacting protein.


Subject(s)
Chitin Synthase/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Membrane Transport Proteins/genetics , Wings, Animal/growth & development , Amino Acid Sequence , Animals , Chitin Synthase/chemistry , Chitin Synthase/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epidermis/growth & development , Larva/genetics , Larva/growth & development , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Phylogeny , Sequence Alignment
19.
Gene ; 809: 146002, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34648919

ABSTRACT

We aimed to explain the reason and function of the successive expression of ecdysone-responsive transcription factors (ERTFs) and related cuticular protein (CP) genes during transformation from larva to pupa. The regulation of the expression of CP genes by ERTFs was examined by in vitro wing disc culture and reporter assay using a gene gun transduction system. Two CP genes that showed expression peaks at different stages-BmorCPG12 at W3L and BmorCPH2 at P0 stage-were selected and examined. Reporter constructs conveying putative BHR3, ßFTZ-F1, BHR39, and E74A binding sites of BmorCPG12 and BmorCPH2 showed promoter activity when introduced into wing discs. In the present study, we showed the functioning of the putative BHR3 and E74A binding sites, together with putative ßFTZ-F1 binding sites, on the activation of CP genes, and different ERTF binding sites functioned in one CP gene. From these, we conclude that BHR3, ßFTZ-F1, and E74A that are successively expressed bring about the successive expression of CP genes, resulting in insect metamorphosis. In addition to this, reporter constructs conveying putative BHR39 binding sites of BmorCPG12 and BmorCPH2 showed negative regulation.


Subject(s)
Bombyx/genetics , Ecdysone/metabolism , Insect Proteins/genetics , Metamorphosis, Biological/genetics , Transcription Factors/genetics , Animals , Binding Sites , Bombyx/physiology , Ecdysone/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Insect Proteins/metabolism , Larva/genetics , Mutagenesis, Site-Directed , Pupa/genetics , Transcription Factors/metabolism , Wings, Animal/growth & development
20.
Dev Biol ; 482: 7-16, 2022 02.
Article in English | MEDLINE | ID: mdl-34822846

ABSTRACT

Regeneration is a response mechanism aiming to reconstruct lost or damaged structures. To achieve this, the cells repopulating the lost tissue often have to change their original identity, a process that involves chromatin remodelling.We have analysed the issue of chromatin remodelling during regeneration in the wing disc of Drosophila . In this disc the ablation of the central region (the pouch) induces the regenerative response of the cells from the lateral region (the hinge), which reconstitute the wing pouch. We have examined euchromatin and heterochromatin histone marks during the process and find that heterochromatin marks disappear but are recovered when regeneration is complete. Euchromatin marks are not modified. We also describe the transcription of two retrotransposons, Roo and F-element in the regenerating cells. We have established a temporal correlation between the alterations of heterochromatin marks and the levels of transcription of two retrotransposons, Roo and F-element, both during embryonic development and in the regeneration process.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Drosophila melanogaster/embryology , Imaginal Discs/growth & development , Regeneration/physiology , Retroelements/genetics , Wings, Animal/embryology , Acetylation , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Euchromatin/metabolism , Heterochromatin/metabolism , Histones/metabolism , Long Interspersed Nucleotide Elements/genetics , Methylation , Wings, Animal/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...