Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.367
Filter
1.
Dev Growth Differ ; 65(6): 348-359, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37310211

ABSTRACT

The acquisition of wings was a key event in insect evolution. As hemimetabolous insects were the first group to acquire functional wings, establishing the mechanisms of wing formation in this group could provide useful insights into their evolution. In this study, we aimed to elucidate the expression and function of the gene scalloped (sd), which is involved in wing formation in Drosophila melanogaster, and in Gryllus bimaculatus mainly during postembryonic development. Expression analysis showed that sd is expressed in the tergal edge, legs, antennae, labrum, and cerci during embryogenesis and in the distal margin of the wing pads from at least the sixth instar in the mid to late stages. Because sd knockout caused early lethality, nymphal RNA interference experiments were performed. Malformations were observed in the wings, ovipositor, and antennae. By analyzing the effects on wing morphology, it was revealed that sd is mainly involved in the formation of the margin, possibly through the regulation of cell proliferation. In conclusion, sd might regulate the local growth of wing pads and influence wing margin morphology in Gryllus.


Subject(s)
Embryonic Development , Gryllidae , Insect Proteins , Transcription Factors , Wings, Animal , Animals , Cell Cycle , Cell Proliferation , Embryonic Development/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Wings, Animal/embryology , Wings, Animal/metabolism , Gryllidae/embryology , Gryllidae/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Development ; 149(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36355083

ABSTRACT

Morphogens of the Hh family trigger gene expression changes in receiving cells in a concentration-dependent manner to regulate their identity, proliferation, death or metabolism, depending on the tissue or organ. This variety of responses relies on a conserved signaling pathway. Its logic includes a negative-feedback loop involving the Hh receptor Ptc. Here, using experiments and computational models we study and compare the different spatial signaling profiles downstream of Hh in several developing Drosophila organs. We show that the spatial distributions of Ptc and the activator transcription factor CiA in wing, antenna and ocellus show similar features, but are markedly different from that in the compound eye. We propose that these two profile types represent two time points along the signaling dynamics, and that the interplay between the spatial displacement of the Hh source in the compound eye and the negative-feedback loop maintains the receiving cells effectively in an earlier stage of signaling. These results show how the interaction between spatial and temporal dynamics of signaling and differentiation processes may contribute to the informational versatility of the conserved Hh signaling pathway.


Subject(s)
Drosophila , Hedgehog Proteins , Signal Transduction , Drosophila/embryology , Animals , Hedgehog Proteins/physiology , Wings, Animal/embryology , Compound Eye, Arthropod/embryology
3.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35088829

ABSTRACT

A long-standing view in the field of evo-devo is that insect forewings develop without any Hox gene input. The Hox gene Antennapedia (Antp), despite being expressed in the thoracic segments of insects, has no effect on wing development. This view has been obtained from studies in two main model species: Drosophila and Tribolium. Here, we show that partial loss of function of Antp resulted in reduced and malformed adult wings in Bombyx, Drosophila and Tribolium. Antp mediates wing growth in Bombyx by directly regulating the ecdysteriod biosynthesis enzyme gene (shade) in the wing tissue, which leads to local production of the growth hormone 20-hydroxyecdysone. Additional targets of Antp are wing cuticular protein genes CPG24, CPH28 and CPG9, which are essential for wing development. We propose, therefore, that insect wing development occurs in an Antp-dependent manner. This article has an associated 'The people behind the papers' interview.


Subject(s)
Homeodomain Proteins/metabolism , Insect Proteins/metabolism , Wings, Animal/embryology , Animals , Bombyx , Drosophila , Ecdysterone/metabolism , Homeodomain Proteins/genetics , Insect Proteins/genetics , Loss of Function Mutation , Morphogenesis , Tribolium , Wings, Animal/metabolism
4.
Cell Rep ; 38(4): 110288, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35081337

ABSTRACT

A fundamental question in biology is how embryonic development is timed between different species. To address this problem, we compared wing development in the quail and the larger chick. We reveal that pattern formation is faster in the quail as determined by the earlier activation of 5'Hox genes, termination of developmental organizers (Shh and Fgf8), and the laying down of the skeleton (Sox9). Using interspecies tissue grafts, we show that developmental timing can be reset during a critical window of retinoic acid signaling. Accordingly, extending the duration of retinoic acid signaling switches developmental timing between the quail and the chick and the chick and the larger turkey. However, the incremental growth rate is comparable between all three species, suggesting that the pace of development primarily governs differences in the expansion of the skeletal pattern. The widespread distribution of retinoic acid could coordinate developmental timing throughout the embryo.


Subject(s)
Embryonic Development/physiology , Embryonic Induction/physiology , Tretinoin/metabolism , Wings, Animal/embryology , Animals , Chick Embryo , Quail/embryology , Turkeys/embryology
5.
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
6.
Elife ; 102021 08 17.
Article in English | MEDLINE | ID: mdl-34402427

ABSTRACT

Spatial boundaries formed during animal development originate from the pre-patterning of tissues by signaling molecules, called morphogens. The accuracy of boundary location is limited by the fluctuations of morphogen concentration that thresholds the expression level of target gene. Producing more morphogen molecules, which gives rise to smaller relative fluctuations, would better serve to shape more precise target boundaries; however, it incurs more thermodynamic cost. In the classical diffusion-depletion model of morphogen profile formation, the morphogen molecules synthesized from a local source display an exponentially decaying concentration profile with a characteristic length λ. Our theory suggests that in order to attain a precise profile with the minimal cost, λ should be roughly half the distance to the target boundary position from the source. Remarkably, we find that the profiles of morphogens that pattern the Drosophila embryo and wing imaginal disk are formed with nearly optimal λ. Our finding underscores the cost-effectiveness of precise morphogen profile formation in Drosophila development.


Subject(s)
Body Patterning , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Models, Biological , Animals , Diffusion , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Signal Transduction , Time Factors , Wings, Animal/embryology , Wings, Animal/metabolism
7.
Elife ; 102021 07 22.
Article in English | MEDLINE | ID: mdl-34292155

ABSTRACT

Morphogen signaling proteins disperse across tissues to activate signal transduction in target cells. We investigated dispersion of Hedgehog (Hh), Wnt homolog Wingless (Wg), and Bone morphogenic protein homolog Decapentaplegic (Dpp) in the Drosophila wing imaginal disc. We discovered that delivery of Hh, Wg, and Dpp to their respective targets is regulated. We found that <5% of Hh and <25% of Wg are taken up by disc cells and activate signaling. The amount of morphogen that is taken up and initiates signaling did not change when the level of morphogen expression was varied between 50 and 200% (Hh) or 50 and 350% (Wg). Similar properties were observed for Dpp. We analyzed an area of 150 µm×150 µm that includes Hh-responding cells of the disc as well as overlying tracheal cells and myoblasts that are also activated by disc-produced Hh. We found that the extent of signaling in the disc was unaffected by the presence or absence of the tracheal and myoblast cells, suggesting that the mechanism that disperses Hh specifies its destinations to particular cells, and that target cells do not take up Hh from a common pool.


Subject(s)
Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Imaginal Discs/metabolism , Signal Transduction , Wnt1 Protein/metabolism , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Morphogenesis , Wings, Animal/embryology , Wnt1 Protein/genetics
8.
Cells Dev ; 165: 203664, 2021 03.
Article in English | MEDLINE | ID: mdl-33993981

ABSTRACT

The Notch signaling pathway is highly conserved and regulates various fundamental development events. Activation of Notch signaling relies on production of the Notch intracellular domain (NICD), which assembles a transcription factor complex to turn on down-stream targets expression. The mastermind (mam) gene encodes an essential co-activator that permits NICD activity in the cell nucleus. During a somatic mosaic screen in Drosophila, an uncharacterized gene l(2)S9998 is identified as a positive regulator of the Notch signaling pathway. Genetic analysis demonstrates that l(2)S9998 functions at the level of transcriptional activation of Notch targets in the signal receiving cells. Whole genome sequencing reveals that l(2)S9998 is a novel allele of the mam gene, which is further confirmed by complementation tests. Along with three molecularly defined transposon insertions isolated from the screen, four mutants of mam are shown to modulate Notch signaling during fly wing development. Our analysis provides additional genetic resources for understanding mam function and Notch signaling regulation.


Subject(s)
Alleles , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genetic Testing , Nuclear Proteins/genetics , Animals , Chromosome Mapping , DNA Transposable Elements/genetics , Mutagenesis, Insertional/genetics , Receptors, Notch/metabolism , Signal Transduction , Wings, Animal/embryology
9.
Elife ; 102021 03 26.
Article in English | MEDLINE | ID: mdl-33769281

ABSTRACT

Tissue organization is often characterized by specific patterns of cell morphology. How such patterns emerge in developing tissues is a fundamental open question. Here, we investigate the emergence of tissue-scale patterns of cell shape and mechanical tissue stress in the Drosophila wing imaginal disc during larval development. Using quantitative analysis of the cellular dynamics, we reveal a pattern of radially oriented cell rearrangements that is coupled to the buildup of tangential cell elongation. Developing a laser ablation method, we map tissue stresses and extract key parameters of tissue mechanics. We present a continuum theory showing that this pattern of cell morphology and tissue stress can arise via self-organization of a mechanical feedback that couples cell polarity to active cell rearrangements. The predictions of this model are supported by knockdown of MyoVI, a component of mechanosensitive feedback. Our work reveals a mechanism for the emergence of cellular patterns in morphogenesis.


During development, carefully choreographed cell movements ensure the creation of a healthy organism. To determine their identity and place across a tissue, cells can read gradients of far-reaching signaling molecules called morphogens; in addition, physical forces can play a part in helping cells acquire the right size and shape. Indeed, cells are tightly attached to their neighbors through connections linked to internal components. Structures or proteins inside the cells can pull on these junctions to generate forces that change the physical features of a cell. However, it is poorly understood how these forces create patterns of cell size and shape across a tissue. Here, Dye, Popovic et al. combined experiments with physical models to examine how cells acquire these physical characteristics across the developing wing of fruit fly larvae. This revealed that cells pushing and pulling on one another create forces that trigger internal biochemical reorganization ­ for instance, force-generating structures become asymmetrical. In turn, the cells exert additional forces on their neighbors, setting up a positive feedback loop which results in cells adopting the right size and shape across the organ. As such, cells in the fly wing can spontaneously self-organize through the interplay of mechanical and biochemical signals, without the need for pre-existing morphogen gradients. A refined understanding of how physical forces shape cells and organs would help to grasp how defects can emerge during development. This knowledge would also allow scientists to better grow tissues and organs in the laboratory, both for theoretical research and regenerative medicine.


Subject(s)
Cell Shape , Drosophila melanogaster/physiology , Imaginal Discs/physiology , Mechanotransduction, Cellular , Wings, Animal/physiology , Animals , Body Patterning , Cell Division , Cell Polarity , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Feedback, Physiological , Female , Imaginal Discs/embryology , Male , Models, Biological , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Stress, Mechanical , Time Factors , Wings, Animal/embryology
10.
Insect Biochem Mol Biol ; 131: 103551, 2021 04.
Article in English | MEDLINE | ID: mdl-33556555

ABSTRACT

Matrix metalloproteinases (MMPs) are the major proteinases that process or degrade numerous extracellular matrix (ECM) components and are evolutionarily conserved from nematodes to humans. During molting in insects, the old cuticle is removed and replaced by a new counterpart. Although the regulatory mechanisms of hormones and nutrients in molting have been well studied, very little is known about the roles of ECM-modifying enzymes in this process. Here, we found that MMPs are necessary for imaginal molting of the American cockroach, Periplaneta americana. Inhibition of Mmp activity via inhibitor treatment led to the failure of eclosion and wing expansion. Five Mmps genes were identified from the P. americana genome, and PaMmp2 played the dominant roles during molting. Further microscopic investigations showed that newly formed adult cuticles were attenuated and that then chitin content was reduced upon Mmp inhibition. Transcriptomic analysis of the integument demonstrated that multiple signaling and metabolic pathways were changed. Microscopic investigation of the wings showed that epithelial cells were restrained together because they were incapable of degrading the ECM upon Mmp inhibition. Transcriptomic analysis of the wing identified dozens of possible genes functioned in wing expansion. This is the first study to show the essential roles of Mmps in the nymph-adult transition of hemimetabolous insects.


Subject(s)
Matrix Metalloproteinases , Periplaneta , Wings, Animal , Animals , Chitin/metabolism , Gene Expression Profiling , Genes, Insect , Larva/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Metamorphosis, Biological , Molting , Nymph/metabolism , Periplaneta/embryology , Periplaneta/genetics , Periplaneta/metabolism , Periplaneta/physiology , Wings, Animal/embryology , Wings, Animal/metabolism
11.
Insect Biochem Mol Biol ; 131: 103552, 2021 04.
Article in English | MEDLINE | ID: mdl-33577967

ABSTRACT

Regeneration is a common phenomenon in various organisms by which tissues restore the damaged or naturally detached parts. In insects, appendage regeneration takes place during the embryonic, larval and pupal stages for individual survival. The wing disc of black cutworm Agrotis ypsilon has the capacity of regeneration after ablation, but understanding of molecular mechanisms in wing disc regeneration is still limited. After ablation of partial or whole wing discs before the fifth instar larval stage, the adult wings appeared to be normal. In the last two larval stages, ablation of the left wing disc led to smaller corresponding adult wing. Cell proliferation was reduced in the ablated wing disc but was gradually recovered two days post ablation. Transcriptome analysis found that genes in the mitogen-activated protein kinase (MAPK) pathway were upregulated. Repression of gene expression in this pathway, including Ras oncogene at 64B (Ras64B), Downstream of raf1 (Dsor1), and cAMP-dependent protein kinase catalytic subunit 3 (Pka-C3) by RNA interference after ablation, led to diminishment of both adult wings, suggesting that the MAPK signaling is essential for wing growth. Additionally, cell proliferation was still decelerated by injecting Ras64B, Dsor, or Pka-C3 dsRNA two days after ablation, indicating that the MAPK signaling-regulated cell proliferation is essential for growth. These results provide molecular clues to the regulation of cell proliferation during regeneration in lepidopteran insects.


Subject(s)
Moths , Regeneration , Wings, Animal , Animals , Cell Proliferation/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Insect , Genes, ras , Larva/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Moths/embryology , Moths/metabolism , Pupa/metabolism , Regeneration/genetics , Regeneration/physiology , Signal Transduction , Wings, Animal/embryology , Wings, Animal/growth & development
12.
Insect Sci ; 28(4): 901-916, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32536018

ABSTRACT

Chitinase degrades chitin in the old epidermis or peritrophic matrix of insects, which ensures normal development and metamorphosis. In our previous work, we comprehensively studied the function of SfCht7 in Sogatella furcifera. However, the number and function of chitinase genes in S. furcifera remain unknown. Here, we identified 12 full-length chitinase transcripts from S. furcifera, which included nine chitinase (Cht), two imaginal disc growth factor (IDGF), and one endo-ß-N-acetylglucosaminidase (ENGase) genes. Expression analysis results revealed that the expression levels of eight genes (SfCht3, SfCht5, SfCht6-1, SfCht6-2, SfCht7, SfCht8, SfCht10, and SfIDGF2) with similar transcript levels peaked prior to molting of each nymph and were highly expressed in the integument. Based on RNA interference (RNAi), description of the functions of each chitinase gene indicated that the silencing of SfCht5, SfCht10, and SfIDGF2 led to molting defects and lethality. RNAi inhibited the expressions of SfCht5, SfCht7, SfCht10, and SfIDGF2, which led to downregulated expressions of chitin synthase 1 (SfCHS1, SfCHS1a, and SfCHS1b) and four chitin deacetylase genes (SfCDA1, SfCDA2, SfCDA3, and SfCDA4), and caused a change in the expression level of two trehalase genes (TRE1 and TRE2). Furthermore, silencing of SfCht7 induced a significant decrease in the expression levels of three wing development-related genes (SfWG, SfDpp, and SfHh). In conclusion, SfCht5, SfCht7, SfCht10, and SfIDGF2 play vital roles in nymph-adult transition and are involved in the regulation of chitin metabolism, and SfCht7 is also involved in wing development; therefore, these genes are potential targets for control of S. furcifera.


Subject(s)
Chitinases/genetics , Hemiptera , Metamorphosis, Biological/genetics , Acetylglucosaminidase/genetics , Animal Shells/embryology , Animal Shells/growth & development , Animals , Gene Expression Regulation, Developmental , Genes, Insect , Hemiptera/embryology , Hemiptera/genetics , Hemiptera/physiology , Imaginal Discs/embryology , Intercellular Signaling Peptides and Proteins/genetics , Molting/genetics , Nymph/growth & development , Nymph/physiology , Wings, Animal/embryology , Wings, Animal/growth & development
13.
Int J Dev Biol ; 65(4-5-6): 357-364, 2021.
Article in English | MEDLINE | ID: mdl-32930350

ABSTRACT

Cell differentiation, proliferation, and morphogenesis are generally driven by instructive signals that are sent and interpreted by adjacent tissues, a process known as induction. Cell recruitment is a particular case of induction in which differentiated cells produce a signal that drives adjacent cells to differentiate into the same type as the inducers. Once recruited, these new cells may become inducers to continue the recruitment process, closing a feed-forward loop that propagates the growth of a specific cell-type population. So far, little attention has been given to cell recruitment as a developmental mechanism. Here, we review the components of cell recruitment and discuss its contribution to development in three different examples: the Drosophila wing, the vertebrate inner ear, and the mammalian thyroid gland. Finally, we posit some open questions about the role of cell recruitment in organ patterning and growth.


Subject(s)
Drosophila , Mammals , Morphogenesis , Vertebrates , Animals , Drosophila/embryology , Ear, Inner/embryology , Gene Expression Regulation, Developmental , Mammals/embryology , Thyroid Gland/embryology , Vertebrates/embryology , Wings, Animal/embryology
14.
Development ; 147(22)2020 11 17.
Article in English | MEDLINE | ID: mdl-33028612

ABSTRACT

Cell extrusion is a crucial regulator of epithelial tissue development and homeostasis. Epithelial cells undergoing apoptosis, bearing pathological mutations or possessing developmental defects are actively extruded toward elimination. However, the molecular mechanisms of Drosophila epithelial cell extrusion are not fully understood. Here, we report that activation of the conserved Hippo (Hpo) signaling pathway induces both apical and basal cell extrusion in the Drosophila wing disc epithelia. We show that canonical Yorkie targets Diap1, Myc and Cyclin E are not required for either apical or basal cell extrusion induced by activation of this pathway. Another target gene, bantam, is only involved in basal cell extrusion, suggesting novel Hpo-regulated apical cell extrusion mechanisms. Using RNA-seq analysis, we found that JNK signaling is activated in the extruding cells. We provide genetic evidence that JNK signaling activation is both sufficient and necessary for Hpo-regulated cell extrusion. Furthermore, we demonstrate that the ETS-domain transcription factor Ets21c, an ortholog of proto-oncogenes FLI1 and ERG, acts downstream of JNK signaling to mediate apical cell extrusion. Our findings reveal a novel molecular link between Hpo signaling and cell extrusion.


Subject(s)
Drosophila Proteins/metabolism , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Signal Transduction/physiology , Wings, Animal/embryology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Imaginal Discs/cytology , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-ets/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Wings, Animal/cytology , YAP-Signaling Proteins
15.
Development ; 147(22)2020 11 17.
Article in English | MEDLINE | ID: mdl-33028613

ABSTRACT

Hedgehog (Hh) is an evolutionarily conserved signaling protein that has essential roles in animal development and homeostasis. We investigated Hh signaling in the region of the Drosophila wing imaginal disc that produces Hh and is near the tracheal air sac primordium (ASP) and myoblasts. Hh distributes in concentration gradients in the anterior compartment of the wing disc, ASP and myoblasts, and activates genes in each tissue. Some targets of Hh signal transduction are common to the disc, ASP and myoblasts, whereas others are tissue-specific. Signaling in the three tissues is cytoneme-mediated and cytoneme-dependent. Some ASP cells project cytonemes that receive both Hh and Branchless (Bnl), and some targets regulated by Hh signaling in the ASP are also dependent on Bnl signal transduction. We conclude that the single source of Hh in the wing disc regulates cell type-specific responses in three discreet target tissues.


Subject(s)
Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Imaginal Discs/metabolism , Signal Transduction , Wings, Animal/embryology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/genetics , Imaginal Discs/cytology , Wings, Animal/cytology
16.
PLoS Comput Biol ; 16(8): e1008105, 2020 08.
Article in English | MEDLINE | ID: mdl-32817654

ABSTRACT

Epithelial sheets define organ architecture during development. Here, we employed an iterative multiscale computational modeling and quantitative experimental approach to decouple direct and indirect effects of actomyosin-generated forces, nuclear positioning, extracellular matrix, and cell-cell adhesion in shaping Drosophila wing imaginal discs. Basally generated actomyosin forces generate epithelial bending of the wing disc pouch. Surprisingly, acute pharmacological inhibition of ROCK-driven actomyosin contractility does not impact the maintenance of tissue height or curved shape. Computational simulations show that ECM tautness provides only a minor contribution to modulating tissue shape. Instead, passive ECM pre-strain serves to maintain the shape independent from actomyosin contractility. These results provide general insight into how the subcellular forces are generated and maintained within individual cells to induce tissue curvature. Thus, the results suggest an important design principle of separable contributions from ECM prestrain and actomyosin tension during epithelial organogenesis and homeostasis.


Subject(s)
Actomyosin/metabolism , Epithelium/anatomy & histology , Extracellular Matrix/metabolism , Animals , Drosophila/anatomy & histology , Drosophila/embryology , Drosophila/metabolism , Epithelium/metabolism , Phosphorylation , Wings, Animal/anatomy & histology , Wings, Animal/embryology , Wings, Animal/metabolism
17.
Int J Dev Biol ; 64(1-2-3): 159-165, 2020.
Article in English | MEDLINE | ID: mdl-32659004

ABSTRACT

Differential specification of dorsal flight appendages, wing and haltere, in Drosophila provides an excellent model system to address a number of important questions in developmental biology at the levels of molecules, pathways, tissues, organs, organisms and evolution. Here we discuss the mechanism by which the Hox protein Ubx recognizes and regulates its downstream targets, implications of the same in growth control at cellular and organ level and finally the evolution of haltere from ancestral hindwings in other holometabolous insects.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Homeodomain Proteins/metabolism , Organogenesis , Transcription Factors/metabolism , Wings, Animal/embryology , Wings, Animal/physiology , Animals , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Transcription Factors/genetics , Wings, Animal/anatomy & histology
18.
Development ; 147(15)2020 08 14.
Article in English | MEDLINE | ID: mdl-32611603

ABSTRACT

Morphogens are important signalling molecules for tissue development and their secretion requires tight regulation. In the wing imaginal disc of flies, the morphogen Wnt/Wingless is apically presented by the secreting cell and re-internalized before final long-range secretion. Why Wnt molecules undergo these trafficking steps and the nature of the regulatory control within the endosomal compartment remain unclear. Here, we have investigated how Wnts are sorted at the level of endosomes by the versatile v-SNARE Ykt6. Using in vivo genetics, proximity-dependent proteomics and in vitro biochemical analyses, we show that most Ykt6 is present in the cytosol, but can be recruited to de-acidified compartments and recycle Wnts to the plasma membrane via Rab4-positive recycling endosomes. Thus, we propose a molecular mechanism by which producing cells integrate and leverage endocytosis and recycling via Ykt6 to coordinate extracellular Wnt levels.


Subject(s)
Drosophila Proteins/metabolism , Endosomes/metabolism , R-SNARE Proteins/metabolism , Wings, Animal/embryology , Wnt Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Endosomes/genetics , Epithelium/embryology , R-SNARE Proteins/genetics , Wnt Proteins/genetics
19.
PLoS Genet ; 16(6): e1008792, 2020 06.
Article in English | MEDLINE | ID: mdl-32579612

ABSTRACT

While rare pathogenic copy-number variants (CNVs) are associated with both neuronal and non-neuronal phenotypes, functional studies evaluating these regions have focused on the molecular basis of neuronal defects. We report a systematic functional analysis of non-neuronal defects for homologs of 59 genes within ten pathogenic CNVs and 20 neurodevelopmental genes in Drosophila melanogaster. Using wing-specific knockdown of 136 RNA interference lines, we identified qualitative and quantitative phenotypes in 72/79 homologs, including 21 lines with severe wing defects and six lines with lethality. In fact, we found that 10/31 homologs of CNV genes also showed complete or partial lethality at larval or pupal stages with ubiquitous knockdown. Comparisons between eye and wing-specific knockdown of 37/45 homologs showed both neuronal and non-neuronal defects, but with no correlation in the severity of defects. We further observed disruptions in cell proliferation and apoptosis in larval wing discs for 23/27 homologs, and altered Wnt, Hedgehog and Notch signaling for 9/14 homologs, including AATF/Aatf, PPP4C/Pp4-19C, and KIF11/Klp61F. These findings were further supported by tissue-specific differences in expression patterns of human CNV genes, as well as connectivity of CNV genes to signaling pathway genes in brain, heart and kidney-specific networks. Our findings suggest that multiple genes within each CNV differentially affect both global and tissue-specific developmental processes within conserved pathways, and that their roles are not restricted to neuronal functions.


Subject(s)
DNA Copy Number Variations , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Neurodevelopmental Disorders/genetics , Animals , Compound Eye, Arthropod/embryology , Compound Eye, Arthropod/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Organ Specificity , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Wings, Animal/embryology , Wings, Animal/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
20.
RNA Biol ; 17(9): 1342-1351, 2020 09.
Article in English | MEDLINE | ID: mdl-32401141

ABSTRACT

microRNAs (miRNA) are small non-coding RNAs that modulate the myriad biological activities by targeting genes, and many studies showed that miRNAs played a pivotal role in insect development. Here, we find that Bm-miRNA (miR-34) controls larval growth and wing morphology by targeting BmE74 and BmCPG4. Overexpression of miR-34 in the whole body caused a smaller body size, partially displays deformed wings and venation defects in adults. Ablation of miR-34 by transgenic CRISPR/Cas9 technology resulted in a severe developmental delay during the larval stage. Moreover, we confirmed that miR-34 directly targeted BmE74 and BmCPG4 by using a dual luciferase reporter assay in HEK293T cells. Remarkably, loss-of-function of BmCPG4 caused wing defects, which was similar to the phenotype of miR-34 overexpression in animals. In addition, our analysis revealed that ecdysone strongly inhibited miR-34 expression in vivo. Taken together, our study identifies miR-34 as a modulator that regulates larval growth and wing morphogenesis by directly modulating ecdysone signalling and cuticle protein in Bombyx mori.


Subject(s)
Bombyx/embryology , Bombyx/genetics , Ecdysone/metabolism , MicroRNAs/genetics , Morphogenesis/genetics , Signal Transduction , Wings, Animal/embryology , Animals , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Larva , Loss of Function Mutation , Organogenesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...