1
|
Li X, Liu M, Xing Y, Niu Y, Liu TH, Sun JL, Liu Y, Hemba-Waduge RUS, Ji JY. Distinct effects of CDK8 module subunits on cellular growth and proliferation in Drosophila. Development 2024; 151:dev203111. [PMID: 39531377 PMCID: PMC11634032 DOI: 10.1242/dev.203111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The Mediator complex plays a pivotal role in facilitating RNA polymerase II-dependent transcription in eukaryotes. Within this complex, the CDK8 kinase module (CKM), comprising CDK8, Cyclin C (CycC), Med12 and Med13, serves as a dissociable subcomplex that modulates the activity of the small Mediator complex. Genetic studies in Drosophila have revealed distinct phenotypes associated with mutations in CKM subunits, but the underlying mechanisms have remained unclear. Using Drosophila as a model, we generated transgenic strains to deplete individually or simultaneously the four CKM subunits in all possible combinations, uncovering unique phenotypes in the eyes and wings. Depletion of CDK8-CycC enhanced E2F1 target gene expression and promoted cell-cycle progression, whereas Med12-Med13 depletion had no significant impact on these processes. Instead, depleting Med12-Med13 altered the expression of ribosomal protein genes and fibrillarin, and reduced nascent protein synthesis, indicating a severe reduction in ribosome biogenesis and cellular growth compared to the loss of CDK8-CycC. These findings reveal distinct in vivo roles for CKM subunits, with Med12-Med13 disruption having a more pronounced effect on ribosome biogenesis and protein synthesis than CDK8-CycC loss.
Collapse
Affiliation(s)
- Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Mengmeng Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Yue Xing
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ye Niu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Tzu-Hao Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Jasmine L. Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Yanwu Liu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Rajitha-Udakara-Sampath Hemba-Waduge
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| |
Collapse
|
2
|
Li X, Liu M, Xing Y, Niu Y, Liu TH, Sun JL, Liu Y, Hemba-Waduge RUS, Ji JY. Distinct effects of CDK8 module subunits on cellular growth and proliferation in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591924. [PMID: 38746212 PMCID: PMC11092604 DOI: 10.1101/2024.04.30.591924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The Mediator complex, composed of about 30 conserved subunits, plays a pivotal role in facilitating RNA polymerase II-dependent transcription in eukaryotes. Within this complex, the CDK8 kinase module (CKM), comprising Med12, Med13, CDK8, and CycC (Cyclin C), serves as a dissociable subcomplex that modulates the activity of the small Mediator complex. Genetic studies in Drosophila have revealed distinct phenotypes of CDK8-CycC and Med12-Med13 mutations, yet the underlying mechanism has remained unknown. Here, using Drosophila as a model organism, we show that depleting CDK8-CycC enhances E2F1 target gene expression and promotes cell-cycle progression. Conversely, depletion of Med12-Med13 affects the expression of ribosomal protein genes and fibrillarin, indicating a more severe reduction in ribosome biogenesis and cellular growth compared to the loss of CDK8-CycC. Moreover, we found that the stability of CDK8 and CycC relies on Med12 and Med13, with a mutually interdependent relationship between Med12 and Med13. Furthermore, CycC stability depends on the other three CKM subunits. These findings reveal distinct roles for CKM subunits in vivo , with Med12-Med13 disruption exerting a more pronounced impact on ribosome biogenesis and cellular growth compared to the loss of CDK8-CycC. Significance The CDK8 kinase module (CKM), comprising CDK8, CycC, Med12, and Med13, is essential in the Mediator complex for RNA polymerase II-dependent transcription in eukaryotes. While expected to function jointly, CKM subunit mutations result in distinct phenotypes in Drosophila . This study investigates the mechanisms driving these differing effects. Our analysis reveals the role of Med12-Med13 pair in regulating ribosomal biogenesis and cellular growth, contrasting with the involvement of CDK8-CycC in E2F1-dependent cell-cycle progression. Additionally, an asymmetric interdependence in the stability of CDK8-CycC and Med12-Med13 was observed. CKM mutations or overexpression are associated with cancers and cardiovascular diseases. Our findings underscore the distinct impacts of CKM mutations on cellular growth and proliferation, advancing our understanding of their diverse consequences in vivo .
Collapse
|
3
|
Dong W, Li T, Wu WJ, Zhang XB. Cell lineage analysis reveals signal tracing and compartment characterisation in Drosophila haltere. INSECT MOLECULAR BIOLOGY 2023; 32:583-591. [PMID: 37212394 DOI: 10.1111/imb.12857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023]
Abstract
Insect halteres, as specialised hind wings, play an important role during aerial manoeuvres. In Drosophila, halteres and wings are homologous appendages with different morphology. Previous studies have focused on the metamorphosis of halteres, while current knowledge about its cell lineage and regional compartmentalization is still limited. In this study, we performed cell-lineage tracing of canonical landmark signals in halteres and present a simple model for haltere development. Cell lineage tracing in wings was used as a reference. The nub showed wing-like expressions in halteres, whereas hth and pnr exhibited different expressions in adult wings and halteres. The lineage tracing revealed that the pouch region gives rise to end-bulb, and hinge cells contribute to proximal haltere formation. Moreover, we demonstrated that twi-expressing cells participate in the cell population of the distal end-bulb. Haematoxylin and eosin staining indicated that muscle cells were present at the distal end-bulb. These results indicated that adult halteres displayed unique cell lineage patterns and the muscle cells are important components of end-bulbs.
Collapse
Affiliation(s)
- Wei Dong
- Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, China
| | - Ting Li
- Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, China
- College of Life Science, Shanxi University, Taiyuan, China
| | - Wen-Jun Wu
- Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, China
- College of Life Science, Shanxi University, Taiyuan, China
| | - Xu-Bo Zhang
- Institute of Applied Biology, Shanxi Key Laboratory of Nucleic Acid Biopesticides, Shanxi University, Taiyuan, China
| |
Collapse
|
4
|
Carmo C, Coelho J, Silva RD, Tavares A, Boavida A, Gaetani P, Guilgur LG, Martinho RG, Oliveira RA. A dual-function SNF2 protein drives chromatid resolution and nascent transcripts removal in mitosis. EMBO Rep 2023; 24:e56463. [PMID: 37462213 PMCID: PMC10481674 DOI: 10.15252/embr.202256463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 09/07/2023] Open
Abstract
Mitotic chromatin is largely assumed incompatible with transcription due to changes in the transcription machinery and chromosome architecture. However, the mechanisms of mitotic transcriptional inactivation and their interplay with chromosome assembly remain largely unknown. By monitoring ongoing transcription in Drosophila early embryos, we reveal that eviction of nascent mRNAs from mitotic chromatin occurs after substantial chromosome compaction and is not promoted by condensin I. Instead, we show that the timely removal of transcripts from mitotic chromatin is driven by the SNF2 helicase-like protein Lodestar (Lds), identified here as a modulator of sister chromatid cohesion defects. In addition to the eviction of nascent transcripts, we uncover that Lds cooperates with Topoisomerase 2 to ensure efficient sister chromatid resolution and mitotic fidelity. We conclude that the removal of nascent transcripts upon mitotic entry is not a passive consequence of cell cycle progression and/or chromosome compaction but occurs via dedicated mechanisms with functional parallelisms to sister chromatid resolution.
Collapse
Affiliation(s)
| | - João Coelho
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Rui D Silva
- Algarve Biomedical Center Research Institute (ABC‐RI) and Faculty of Medicine and Biomedical Sciences (FMCB)Universidade do AlgarveFaroPortugal
| | | | - Ana Boavida
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Present address:
Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle RicercheNaplesItaly
| | | | | | - Rui Gonçalo Martinho
- Algarve Biomedical Center Research Institute (ABC‐RI) and Faculty of Medicine and Biomedical Sciences (FMCB)Universidade do AlgarveFaroPortugal
- Department of Medical Sciences (DCM) and Institute for Biomedicine (iBiMED)Universidade de AveiroAveiroPortugal
| | - Raquel A Oliveira
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Católica Biomedical Research Centre, Católica Medical SchoolUniversidade Católica PortuguesaLisbonPortugal
| |
Collapse
|
5
|
Zhu Y, Li C, Shuai R, Huang Z, Chen F, Wang Y, Zhou Q, Chen J. Experimental study of the mechanism of induction of conjunctival goblet cell hyperexpression using CHIR-99021 in vitro. Biochem Biophys Res Commun 2023; 668:104-110. [PMID: 37245290 DOI: 10.1016/j.bbrc.2023.05.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/15/2023] [Indexed: 05/30/2023]
Abstract
A component of the tear film, mucin is produced by conjunctival goblet cells and is crucial to preserving the tear film's stability. Severe thermal burns, chemical burns, and severe ocular surface diseases can cause extensive damage to the conjunctiva, destroy the secretory function of goblet cells, and affect the stability of the tear film and integrity of the ocular surface. Currently, the expansion efficiency of goblet cells in vitro is low. In this study, we observed that rabbit conjunctival epithelial cells exhibited dense colony morphology after stimulation with the Wnt/β-catenin signaling pathway activator CHIR-99021 and promoted the differentiation of conjunctival goblet cells and the expression of its specific marker Muc5ac, among which the best induction effect was observed after 72 h in vitro culture with 5 μmol/L CHIR-99021. Under optimal culture conditions, CHIR-99021 increased the expression levels of the Wnt/β-catenin signaling pathway factors Frzb, β-catenin, SAM pointed domain containing ETS transcription factor, and glycogen synthase kinase-3β and the levels of the Notch signaling pathway factors Notch1 and Krüppel-like factor 4 while decreasing the expression levels of Jagged-1 and Hes1. The expression level of ABCG2, a marker of epithelial stem cells, was raised to keep rabbit conjunctival epithelial cells from self-renewing. Our study showed that CHIR-99021 stimulation successfully activated the Wnt/β-catenin signaling pathway and conjunctival goblet cell differentiation was stimulated, in which the Notch signaling pathway played a combined role. Those results provide a novel idea for the expansion of goblet cells in vitro.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Chaoqun Li
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ruixue Shuai
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ziqing Huang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fangyuan Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yingwei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Qing Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Jian Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
6
|
Diaz-Torres E, Muñoz-Nava LM, Nahmad M. Coupling cell proliferation rates to the duration of recruitment controls final size of the Drosophila wing. Proc Biol Sci 2022; 289:20221167. [PMID: 36476003 PMCID: PMC9554725 DOI: 10.1098/rspb.2022.1167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Organ growth driven by cell proliferation is an exponential process. As a result, even small variations in proliferation rates, when integrated over a relatively long developmental time, will lead to large differences in size. How organs robustly control their final size despite perturbations in cell proliferation rates throughout development is a long-standing question in biology. Using a mathematical model, we show that in the developing wing of the fruit fly, Drosophila melanogaster, variations in proliferation rates of wing-committed cells are inversely proportional to the duration of cell recruitment, a differentiation process in which a population of undifferentiated cells adopt the wing fate by expressing the selector gene, vestigial. A time-course experiment shows that vestigial-expressing cells increase exponentially while recruitment takes place, but slows down when recruitable cells start to vanish, suggesting that undifferentiated cells may be driving proliferation of wing-committed cells. When this observation is incorporated in our model, we show that the duration of cell recruitment robustly determines a final wing size even when cell proliferation rates of wing-committed cells are perturbed. Finally, we show that this control mechanism fails when perturbations in proliferation rates affect both wing-committed and recruitable cells, providing an experimentally testable hypothesis of our model.
Collapse
Affiliation(s)
- Elizabeth Diaz-Torres
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
7
|
A single WNT enhancer drives specification and regeneration of the Drosophila wing. Nat Commun 2022; 13:4794. [PMID: 35995781 PMCID: PMC9395397 DOI: 10.1038/s41467-022-32400-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Wings have provided an evolutionary advantage to insects and have allowed them to diversify. Here, we have identified in Drosophila a highly robust regulatory mechanism that ensures the specification and growth of the wing not only during normal development but also under stress conditions. We present evidence that a single wing-specific enhancer in the wingless gene is used in two consecutive developmental stages to first drive wing specification and then contribute to mediating the remarkable regenerative capacity of the developing wing upon injury. We identify two evolutionary conserved cis-regulatory modules within this enhancer that are utilized in a redundant manner to mediate these two activities through the use of distinct molecular mechanisms. Whereas Hedgehog and EGFR signalling regulate Wingless expression in early primordia, thus inducing wing specification from body wall precursors, JNK activation in injured tissues induce Wingless expression to promote compensatory proliferation. These results point to evolutionarily linked conservation of wing specification and regeneration to ensure robust development of the wing, perhaps the most relevant evolutionary novelty in insects. The wing is a remarkable evolutionary novelty in insects. Here the authors demonstrate that the specification and regenerative capacity of the wing relies on a single wing-specific enhancer of the wingless gene in Drosophila.
Collapse
|
8
|
Beaven R, Denholm B. Early patterning followed by tissue growth establishes distal identity in Drosophila Malpighian tubules. Front Cell Dev Biol 2022; 10:947376. [PMID: 36060795 PMCID: PMC9437309 DOI: 10.3389/fcell.2022.947376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/28/2022] [Indexed: 12/03/2022] Open
Abstract
Specification and elaboration of proximo-distal (P-D) axes for structures or tissues within a body occurs secondarily from that of the main axes of the body. Our understanding of the mechanism(s) that pattern P-D axes is limited to a few examples such as vertebrate and invertebrate limbs. Drosophila Malpighian/renal tubules (MpTs) are simple epithelial tubules, with a defined P-D axis. How this axis is patterned is not known, and provides an ideal context to understand patterning mechanisms of a secondary axis. Furthermore, epithelial tubules are widespread, and their patterning is not well understood. Here, we describe the mechanism that establishes distal tubule and show this is a radically different mechanism to that patterning the proximal MpT. The distal domain is patterned in two steps: distal identity is specified in a small group of cells very early in MpT development through Wingless/Wnt signalling. Subsequently, this population is expanded by proliferation to generate the distal MpT domain. This mechanism enables distal identity to be established in the tubule in a domain of cells much greater than the effective range of Wingless.
Collapse
Affiliation(s)
| | - Barry Denholm
- Deanery of Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Milán M. Wing regeneration: Single-cell analysis sheds new light. Curr Biol 2022; 32:R842-R844. [PMID: 35944485 DOI: 10.1016/j.cub.2022.06.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The developing wing primordium of Drosophila displays a remarkable capacity to regenerate in response to different types of damage. A new study shows that this capacity relies on the activation of a pro-regenerative gene regulatory network in two distinct cell populations within the blastema.
Collapse
Affiliation(s)
- Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
10
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
11
|
Marcetteau J, Matusek T, Luton F, Thérond PP. Arf6 is necessary for senseless expression in response to wingless signalling during Drosophila wing development. Biol Open 2021; 10:273443. [PMID: 34779478 PMCID: PMC8656404 DOI: 10.1242/bio.058892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022] Open
Abstract
Wnt signalling is a core pathway involved in a wide range of developmental processes throughout the metazoa. In vitro studies have suggested that the small GTP binding protein Arf6 regulates upstream steps of Wnt transduction, by promoting the phosphorylation of the Wnt co-receptor, LRP6, and the release of β-catenin from the adherens junctions. To assess the relevance of these previous findings in vivo, we analysed the consequence of the absence of Arf6 activity on Drosophila wing patterning, a developmental model of Wnt/Wingless signalling. We observed a dominant loss of wing margin bristles and Senseless expression in Arf6 mutant flies, phenotypes characteristic of a defect in high level Wingless signalling. In contrast to previous findings, we show that Arf6 is required downstream of Armadillo/β-catenin stabilisation in Wingless signal transduction. Our data suggest that Arf6 modulates the activity of a downstream nuclear regulator of Pangolin activity in order to control the induction of high level Wingless signalling. Our findings represent a novel regulatory role for Arf6 in Wingless signalling.
Collapse
Affiliation(s)
- Julien Marcetteau
- Université Côte d'Azur; UMR7277 CNRS; Inserm 1091; Institut de Biologie de Valrose (iBV); Parc Valrose, 06108 Nice cedex 2, Nice, France
| | - Tamàs Matusek
- Université Côte d'Azur; UMR7277 CNRS; Inserm 1091; Institut de Biologie de Valrose (iBV); Parc Valrose, 06108 Nice cedex 2, Nice, France
| | - Frédéric Luton
- Université Côte d'Azur; UMR7275 CNRS; Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 660 Route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - Pascal P Thérond
- Université Côte d'Azur; UMR7277 CNRS; Inserm 1091; Institut de Biologie de Valrose (iBV); Parc Valrose, 06108 Nice cedex 2, Nice, France
| |
Collapse
|
12
|
Mehta S, Hingole S, Chaudhary V. The Emerging Mechanisms of Wnt Secretion and Signaling in Development. Front Cell Dev Biol 2021; 9:714746. [PMID: 34485301 PMCID: PMC8415634 DOI: 10.3389/fcell.2021.714746] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/20/2021] [Indexed: 12/22/2022] Open
Abstract
Wnts are highly-conserved lipid-modified secreted proteins that activate multiple signaling pathways. These pathways regulate crucial processes during various stages of development and maintain tissue homeostasis in adults. One of the most fascinating aspects of Wnt protein is that despite being hydrophobic, they are known to travel several cell distances in the extracellular space. Research on Wnts in the past four decades has identified several factors and uncovered mechanisms regulating their expression, secretion, and mode of extracellular travel. More recently, analyses on the importance of Wnt protein gradients in the growth and patterning of developing tissues have recognized the complex interplay of signaling mechanisms that help in maintaining tissue homeostasis. This review aims to present an overview of the evidence for the various modes of Wnt protein secretion and signaling and discuss mechanisms providing precision and robustness to the developing tissues.
Collapse
Affiliation(s)
| | | | - Varun Chaudhary
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| |
Collapse
|
13
|
Tian Y, Smith-Bolton RK. Regulation of growth and cell fate during tissue regeneration by the two SWI/SNF chromatin-remodeling complexes of Drosophila. Genetics 2021; 217:1-16. [PMID: 33683366 DOI: 10.1093/genetics/iyaa028] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/10/2020] [Indexed: 11/12/2022] Open
Abstract
To regenerate, damaged tissue must heal the wound, regrow to the proper size, replace the correct cell types, and return to the normal gene-expression program. However, the mechanisms that temporally and spatially control the activation or repression of important genes during regeneration are not fully understood. To determine the role that chromatin modifiers play in regulating gene expression after tissue damage, we induced ablation in Drosophila melanogaster imaginal wing discs, and screened for chromatin regulators that are required for epithelial tissue regeneration. Here, we show that many of these genes are indeed important for promoting or constraining regeneration. Specifically, the two SWI/SNF chromatin-remodeling complexes play distinct roles in regulating different aspects of regeneration. The PBAP complex regulates regenerative growth and developmental timing, and is required for the expression of JNK signaling targets and the growth promoter Myc. By contrast, the BAP complex ensures correct patterning and cell fate by stabilizing the expression of the posterior gene engrailed. Thus, both SWI/SNF complexes are essential for proper gene expression during tissue regeneration, but they play distinct roles in regulating growth and cell fate.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel K Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
14
|
Ewen-Campen B, Comyn T, Vogt E, Perrimon N. No Evidence that Wnt Ligands Are Required for Planar Cell Polarity in Drosophila. Cell Rep 2021; 32:108121. [PMID: 32905771 PMCID: PMC7641474 DOI: 10.1016/j.celrep.2020.108121] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/04/2020] [Accepted: 08/18/2020] [Indexed: 11/05/2022] Open
Abstract
The frizzled (fz) and dishevelled (dsh) genes are highly conserved members of both the planar cell polarity (PCP) pathway and the Wnt signaling pathway. Given these dual functions, several studies have examined whether Wnt ligands provide a tissue-scale orientation cue for PCP establishment during development, and these studies have reached differing conclusions. Here, we re-examine this issue in the Drosophila melanogaster wing and notum using split-Gal4 co-expression analysis, multiplex somatic CRISPR, and double RNAi experiments. Pairwise loss-of-function experiments targeting wg together with other Wnt genes, via somatic CRISPR or RNAi, do not produce PCP defects in the wing or notum. In addition, somatic CRISPR against evi (aka wntless), which is required for the secretion of Wnt ligands, did not produce detectable PCP phenotypes. Altogether, our results do not support the hypothesis that Wnt ligands contribute to PCP signaling in the Drosophila wing or notum. Previous studies have come to differing conclusions on whether Wnt ligands provide a tissue-level orientation cue for the planar cell polarity pathway. Ewen-Campen et al. re-examine this question in Drosophila using multiplex in vivo CRISPR and double RNAi against Wnt ligands and find no evidence that Wnts are required for PCP patterning.
Collapse
Affiliation(s)
- Ben Ewen-Campen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Typhaine Comyn
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Vogt
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Zecca M, Struhl G. A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 2021; 19:e3001111. [PMID: 33657096 PMCID: PMC8148325 DOI: 10.1371/journal.pbio.3001111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/25/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Development of the Drosophila wing-a paradigm of organ development-is governed by 2 morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Both proteins are produced by defined subpopulations of cells and spread outwards, forming gradients that control gene expression and cell pattern as a function of concentration. They also control growth, but how is unknown. Most studies have focused on Dpp and yielded disparate models in which cells throughout the wing grow at similar rates in response to the grade or temporal change in Dpp concentration or to the different amounts of Dpp "equalized" by molecular or mechanical feedbacks. In contrast, a model for Wg posits that growth is governed by a progressive expansion in morphogen range, via a mechanism in which a minimum threshold of Wg sustains the growth of cells within the wing and recruits surrounding "pre-wing" cells to grow and enter the wing. This mechanism depends on the capacity of Wg to fuel the autoregulation of vestigial (vg)-the selector gene that specifies the wing state-both to sustain vg expression in wing cells and by a feed-forward (FF) circuit of Fat (Ft)/Dachsous (Ds) protocadherin signaling to induce vg expression in neighboring pre-wing cells. Here, we have subjected Dpp to the same experimental tests used to elucidate the Wg model and find that it behaves indistinguishably. Hence, we posit that both morphogens act together, via a common mechanism, to control wing growth as a function of morphogen range.
Collapse
Affiliation(s)
- Myriam Zecca
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
16
|
Won JH, Cho KO. Wg secreted by conventional Golgi transport diffuses and forms Wg gradient whereas Wg tethered to extracellular vesicles do not diffuse. Cell Death Differ 2020; 28:1013-1025. [PMID: 33028960 DOI: 10.1038/s41418-020-00632-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Wingless (Wg)/Wnt family proteins are essential for animal development and adult homeostasis. Drosophila Wg secreted from the dorsal-ventral (DV) midline in wing discs forms a concentration gradient that is shaped by diffusion rate and stability of Wg. To understand how the gradient of extracellular Wg is generated, we compared the secretion route of NRT-Wg, an artificial membrane-tethered form of Wg that is supposedly not secreted but still supports fly development, to that of wild-type Wg. We found that wild-type Wg is secreted by both conventional Golgi transport and via extracellular vesicles (EVs), and NRT-Wg can be also secreted via EVs. Furthermore, wild-type Wg secreted by Golgi transport diffused and formed Wg gradient but Wg-containing EVs did not diffuse at all. In case of Wg stability, Sol narae (Sona), a metalloprotease that cleaves Wg, contributes to generate a steep Wg gradient. Interestingly, Wg was also produced in the presumptive wing blade region, which indicates that NRT-Wg on EVs expressed in the blade allows the blade cells to proliferate and differentiate without Wg diffused from the DV midline. We propose that EV-associated Wg induces Wg signaling in autocrine and juxtaposed manners whereas Wg secreted by Golgi transport forms gradient and acts in the long-range signaling, and different organs differentially utilize these two types of Wg signaling for their own development.
Collapse
Affiliation(s)
- Jong-Hoon Won
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Kyung-Ok Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
17
|
Yu JJS, Maugarny-Calès A, Pelletier S, Alexandre C, Bellaiche Y, Vincent JP, McGough IJ. Frizzled-Dependent Planar Cell Polarity without Secreted Wnt Ligands. Dev Cell 2020; 54:583-592.e5. [PMID: 32888416 PMCID: PMC7497783 DOI: 10.1016/j.devcel.2020.08.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/07/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022]
Abstract
Planar cell polarity (PCP) organizes the orientation of cellular protrusions and migratory activity within the tissue plane. PCP establishment involves the subcellular polarization of core PCP components. It has been suggested that Wnt gradients could provide a global cue that coordinates local PCP with tissue axes. Here, we dissect the role of Wnt ligands in the orientation of hairs of Drosophila wings, an established system for the study of PCP. We found that PCP was normal in quintuple mutant wings that rely solely on the membrane-tethered Wingless for Wnt signaling, suggesting that a Wnt gradient is not required. We then used a nanobody-based approach to trap Wntless in the endoplasmic reticulum, and hence prevent all Wnt secretion, specifically during the period of PCP establishment. PCP was still established. We conclude that, even though Wnt ligands could contribute to PCP, they are not essential, and another global cue must exist for tissue-wide polarization.
Collapse
Affiliation(s)
| | - Aude Maugarny-Calès
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | - Yohanns Bellaiche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | | |
Collapse
|
18
|
Muscle development : a view from adult myogenesis in Drosophila. Semin Cell Dev Biol 2020; 104:39-50. [DOI: 10.1016/j.semcdb.2020.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/17/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
|
19
|
Temporal flexibility of gene regulatory network underlies a novel wing pattern in flies. Proc Natl Acad Sci U S A 2020; 117:11589-11596. [PMID: 32393634 PMCID: PMC7261121 DOI: 10.1073/pnas.2002092117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Developmental genes can be coopted to generate evolutionary novelties by changing their spatial regulation. However, developmental genes seldom act independently, but rather work in a gene regulatory network (GRN). How is it possible to recruit a single gene from a whole GRN? What are the properties that allow parallel cooptions of the same genes during evolution? Here, we show that a novel engrailed gene expression underlies a novel wing color pattern in flies. We show that cooption is facilitated 1) because of GRN flexibility over development and 2) because every single gene of the GRN has its own functional time window. We suggest these two temporal properties could explain why the same gene can be independently recruited several times during evolution. Organisms have evolved endless morphological, physiological, and behavioral novel traits during the course of evolution. Novel traits were proposed to evolve mainly by orchestration of preexisting genes. Over the past two decades, biologists have shown that cooption of gene regulatory networks (GRNs) indeed underlies numerous evolutionary novelties. However, very little is known about the actual GRN properties that allow such redeployment. Here we have investigated the generation and evolution of the complex wing pattern of the fly Samoaia leonensis. We show that the transcription factor Engrailed is recruited independently from the other players of the anterior–posterior specification network to generate a new wing pattern. We argue that partial cooption is made possible because 1) the anterior–posterior specification GRN is flexible over time in the developing wing and 2) this flexibility results from the fact that every single gene of the GRN possesses its own functional time window. We propose that the temporal flexibility of a GRN is a general prerequisite for its possible cooption during the course of evolution.
Collapse
|
20
|
Sui L, Dahmann C. Wingless counteracts epithelial folding by increasing mechanical tension at basal cell edges in Drosophila. Development 2020; 147:147/5/dev184713. [DOI: 10.1242/dev.184713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/28/2020] [Indexed: 01/21/2023]
Abstract
ABSTRACT
The modulation of mechanical tension is important for sculpturing tissues during animal development, yet how mechanical tension is controlled remains poorly understood. In Drosophila wing discs, the local reduction of mechanical tension at basal cell edges results in basal relaxation and the formation of an epithelial fold. Here, we show that Wingless, which is expressed next to this fold, promotes basal cell edge tension to suppress the formation of this fold. Ectopic expression of Wingless blocks fold formation, whereas the depletion of Wingless increases fold depth. Moreover, local depletion of Wingless in a region where Wingless signal transduction is normally high results in ectopic fold formation. The depletion of Wingless also results in decreased basal cell edge tension and basal cell area relaxation. Conversely, the activation of Wingless signal transduction leads to increased basal cell edge tension and basal cell area constriction. Our results identify the Wingless signal transduction pathway as a crucial modulator of mechanical tension that is important for proper wing disc morphogenesis.
Collapse
Affiliation(s)
- Liyuan Sui
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
21
|
Shadkhoo S, Mani M. The role of intracellular interactions in the collective polarization of tissues and its interplay with cellular geometry. PLoS Comput Biol 2019; 15:e1007454. [PMID: 31770364 PMCID: PMC6903760 DOI: 10.1371/journal.pcbi.1007454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/10/2019] [Accepted: 10/01/2019] [Indexed: 11/18/2022] Open
Abstract
Planar cell polarity (PCP), the long-range in-plane polarization of epithelial tissues, provides directional information that guides a multitude of developmental processes at cellular and tissue levels. While it is manifest that cells utilize both intracellular and intercellular interactions, the coupling between the two modules, essential to the coordination of collective polarization, remains an active area of investigation. We propose a generalized reaction-diffusion model to study the role of intracellular interactions in the emergence of long-range polarization, and show that the nonlocality of cytoplasmic interactions, i.e. coupling of membrane proteins localized on different cell-cell junctions, is of vital importance to the faithful detection of weak directional signals, and becomes increasingly more crucial to the stability of polarization against the deleterious effects of large geometric irregularities. We demonstrate that nonlocal interactions are necessary for geometric information to become accessible to the PCP components. The prediction of the model regarding polarization in elongated tissues, is shown to be in agreement with experimental observations, where the polarity emerges perpendicular to the axis of elongation. Core PCP is adopted as a model pathway, in term of which we interpret the model parameters. To this end, we introduce three distinct classes of mutations, (I) in membrane proteins, (II) in cytoplasmic proteins, and (III) local enhancement of geometric disorder. Comparing the in silico and in vivo phenotypes, we show that our model successfully recapitulates the salient phenotypic features of these mutations. Exploring the parameter space helps us shed light on the role of cytoplasmic proteins in cell-cell communications, and make falsifiable predictions regarding the cooperation of cytoplasmic and membrane proteins in the establishment of long-range polarization. Planar cell polarity (PCP) is an indispensable and conserved pathway in morphogenesis. In spite of the advances in understanding the different modules of PCP, a comprehensive picture of the intracellular protein-protein interactions necessary for the emergence of long-range tissue polarity is still lacking. In order to address this question, we devised a generalized reaction-diffusion model, through which we investigated the role of cytoplasmic interactions in PCP pathways. The length scale of intracellular interactions is demonstrated to be crucial to the stability of the cytoplasmic segregation of membrane proteins in disordered tissues, as well as the capacity of polarization field for detecting the gradient and geometrical cues. Finally, three classes of mutants are investigated within the context of our model. Comparison with the in vivo observations allows us to infer the major contributions of cytoplasmic proteins to the emergence of tissue polarity, and make testable predictions regarding the cooperation of cytoplasmic and membrane proteins in the coordination of collective polarization.
Collapse
Affiliation(s)
- Shahriar Shadkhoo
- Kavli Institute for Theoretical Physics, University of California, Santa Barbara, California, United States of America.,Physics Department, University of California, Santa Barbara, California, United States of America
| | - Madhav Mani
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois, United States of America.,NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, Illinois, United States of America.,Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
22
|
McCulloch GA, Oliphant A, Dearden PK, Veale AJ, Ellen CW, Waters JM. Comparative transcriptomic analysis of a wing-dimorphic stonefly reveals candidate wing loss genes. EvoDevo 2019; 10:21. [PMID: 31516688 PMCID: PMC6728979 DOI: 10.1186/s13227-019-0135-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/24/2019] [Indexed: 01/28/2023] Open
Abstract
Background The genetic basis of wing development has been well characterised for model insect species, but remains poorly understood in phylogenetically divergent, non-model taxa. Wing-polymorphic insect species potentially provide ideal systems for unravelling the genetic basis of secondary wing reduction. Stoneflies (Plecoptera) represent an anciently derived insect assemblage for which the genetic basis of wing polymorphism remains unclear. We undertake quantitative RNA-seq of sympatric full-winged versus vestigial-winged nymphs of a widespread wing-dimorphic New Zealand stonefly, Zelandoperla fenestrata, to identify genes potentially involved in wing development and secondary wing loss. Results Our analysis reveals substantial differential expression of wing-development genes between full-winged versus vestigial-winged stonefly ecotypes. Specifically, of 23 clusters showing significant similarity to Drosophila wing development-related genes and their pea aphid orthologues, nine were significantly upregulated in full-winged stonefly ecotypes, whereas only one cluster (teashirt) was substantially upregulated in the vestigial-winged ecotype. Conclusions These findings suggest remarkable conservation of key wing-development pathways throughout 400 Ma of insect evolution. The finding that two Juvenile Hormone pathway clusters were significantly upregulated in vestigial-winged Zelandoperla supports the hypothesis that Juvenile Hormone may play a key role in modulating insect wing polymorphism, as has previously been suggested for other insect lineages.
Collapse
Affiliation(s)
- Graham A McCulloch
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Andrew Oliphant
- 2Genomics Aotearoa and Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Peter K Dearden
- 2Genomics Aotearoa and Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Andrew J Veale
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand.,3Landcare Research, Private Bag 92170, Auckland Mail Centre, Auckland, 1142 New Zealand
| | - Charles W Ellen
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Jonathan M Waters
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| |
Collapse
|
23
|
Chaudhary V, Hingole S, Frei J, Port F, Strutt D, Boutros M. Robust Wnt signaling is maintained by a Wg protein gradient and Fz2 receptor activity in the developing Drosophila wing. Development 2019; 146:dev174789. [PMID: 31399474 PMCID: PMC6703709 DOI: 10.1242/dev.174789] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 07/10/2019] [Indexed: 12/19/2022]
Abstract
Wnts are secreted proteins that regulate cell fate during development of all metazoans. Wnt proteins were proposed to spread over several cells to activate signaling directly at a distance. In the Drosophila wing epithelium, an extracellular gradient of the Wnt1 homolog Wingless (Wg) was observed extending over several cells away from producing cells. Surprisingly, however, it was also shown that a membrane-tethered Neurotactin-Wg fusion protein (NRT-Wg) can largely replace endogenous Wg, leading to proper patterning of the wing. Therefore, the functional range of Wg and whether Wg spreading is required for correct tissue patterning remains controversial. Here, by capturing secreted Wg on cells away from the source, we show that Wg acts over a distance of up to 11 cell diameters to induce signaling. Furthermore, cells located outside the reach of extracellular Wg depend on the Frizzled2 receptor to maintain signaling. Frizzled2 expression is increased in the absence of Wg secretion and is required to maintain signaling and cell survival in NRT-wg wing discs. Together, these results provide insight into the mechanisms by which robust Wnt signaling is achieved in proliferating tissues.
Collapse
Affiliation(s)
- Varun Chaudhary
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
- Department of Biological Science, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, India
| | - Swapnil Hingole
- Department of Biological Science, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, India
| | - Jana Frei
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Fillip Port
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics and Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Abstract
Wnt/Wingless (Wg) signaling controls many aspects of animal development and is deregulated in different human cancers. The transcription factor dTcf/Pangolin (Pan) is the final effector of the Wg pathway in Drosophila and has a dual role in regulating the expression of Wg target genes. In the presence of Wg, dTcf/Pan interacts with β-catenin/Armadillo (Arm) and induces the transcription of Wg targets. In absence of Wg, dTcf/Pan partners with the transcriptional corepressor TLE/Groucho (Gro) and inhibits gene expression. Here, we use the wing imaginal disk of Drosophila as a model to examine the functions that dTcf/Pan plays in a proliferating epithelium. We report a function of dTcf/Pan in growth control and tumorigenesis. Our results show that dTcf/Pan can limit tissue growth in normal development and suppresses tumorigenesis in the context of oncogene up-regulation. We identify the conserved transcription factors Sox box protein 15 (Sox15) and Ftz transcription factor 1 (Ftz-f1) as genes controlled by dTcf/Pan involved in tumor development. In conclusion, this study reports a role for dTcf/Pan as a repressor of normal and oncogenic growth and identifies the genes inducing tumorigenesis downstream of dTcf/Pan.
Collapse
|
25
|
Ruiz-Losada M, Blom-Dahl D, Córdoba S, Estella C. Specification and Patterning of Drosophila Appendages. J Dev Biol 2018; 6:jdb6030017. [PMID: 30011921 PMCID: PMC6162442 DOI: 10.3390/jdb6030017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023] Open
Abstract
Appendages are external projections of the body that serve the animal for locomotion, feeding, or environment exploration. The appendages of the fruit fly Drosophilamelanogaster are derived from the imaginal discs, epithelial sac-like structures specified in the embryo that grow and pattern during larva development. In the last decades, genetic and developmental studies in the fruit fly have provided extensive knowledge regarding the mechanisms that direct the formation of the appendages. Importantly, many of the signaling pathways and patterning genes identified and characterized in Drosophila have similar functions during vertebrate appendage development. In this review, we will summarize the genetic and molecular mechanisms that lead to the specification of appendage primordia in the embryo and their posterior patterning during imaginal disc development. The identification of the regulatory logic underlying appendage specification in Drosophila suggests that the evolutionary origin of the insect wing is, in part, related to the development of ventral appendages.
Collapse
Affiliation(s)
- Mireya Ruiz-Losada
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - David Blom-Dahl
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Sergio Córdoba
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| |
Collapse
|
26
|
Next-generation CRISPR/Cas9 transcriptional activation in Drosophila using flySAM. Proc Natl Acad Sci U S A 2018; 115:4719-4724. [PMID: 29666231 DOI: 10.1073/pnas.1800677115] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CRISPR/Cas9-based transcriptional activation (CRISPRa) has recently emerged as a powerful and scalable technique for systematic overexpression genetic analysis in Drosophila melanogaster We present flySAM, a potent tool for in vivo CRISPRa, which offers major improvements over existing strategies in terms of effectiveness, scalability, and ease of use. flySAM outperforms existing in vivo CRISPRa strategies and approximates phenotypes obtained using traditional Gal4-UAS overexpression. Moreover, because flySAM typically requires only a single sgRNA, it dramatically improves scalability. We use flySAM to demonstrate multiplexed CRISPRa, which has not been previously shown in vivo. In addition, we have simplified the experimental use of flySAM by creating a single vector encoding both the UAS:Cas9-activator and the sgRNA, allowing for inducible CRISPRa in a single genetic cross. flySAM will replace previous CRISPRa strategies as the basis of our growing genome-wide transgenic overexpression resource, TRiP-OE.
Collapse
|
27
|
Worley MI, Alexander LA, Hariharan IK. CtBP impedes JNK- and Upd/STAT-driven cell fate misspecifications in regenerating Drosophila imaginal discs. eLife 2018; 7:30391. [PMID: 29372681 PMCID: PMC5823544 DOI: 10.7554/elife.30391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/19/2018] [Indexed: 12/27/2022] Open
Abstract
Regeneration following tissue damage often necessitates a mechanism for cellular re-programming, so that surviving cells can give rise to all cell types originally found in the damaged tissue. This process, if unchecked, can also generate cell types that are inappropriate for a given location. We conducted a screen for genes that negatively regulate the frequency of notum-to-wing transformations following genetic ablation and regeneration of the wing pouch, from which we identified mutations in the transcriptional co-repressor C-terminal Binding Protein (CtBP). When CtBP function is reduced, ablation of the pouch can activate the JNK/AP-1 and JAK/STAT pathways in the notum to destabilize cell fates. Ectopic expression of Wingless and Dilp8 precede the formation of the ectopic pouch, which is subsequently generated by recruitment of both anterior and posterior cells near the compartment boundary. Thus, CtBP stabilizes cell fates following damage by opposing the destabilizing effects of the JNK/AP-1 and JAK/STAT pathways.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Larissa A Alexander
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
28
|
Sander M, Eichenlaub T, Herranz H. Oncogenic cooperation between Yorkie and the conserved microRNA miR-8 in the wing disc of Drosophila. Development 2018; 145:dev.153817. [DOI: 10.1242/dev.153817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/05/2018] [Indexed: 12/22/2022]
Abstract
Tissue growth has to be carefully controlled to generate well-functioning organs. microRNAs are small noncoding RNAs that modulate the activity of target genes and play a pivotal role in animal development. Understanding the functions of microRNAs in development requires the identification of their target genes. Here, we find that the conserved microRNA miR-8/miR-200 controls tissue growth and homeostasis in the Drosophila wing imaginal disc. Upregulation of miR-8 causes the repression of Yorkie, the effector of the Hippo pathway in Drosophila, and reduces tissue size. Remarkably, coexpression of Yorkie and miR-8 causes the formation of neoplastic tumors. We show that upregulation of miR-8 represses the growth inhibitor brinker, and depletion of brinker cooperates with Yorkie in the formation of neoplastic tumors. Hence, miR-8 modulates a positive growth regulator, Yorkie, and a negative growth regulator, brinker. Deregulation of this network can result in the loss of tissue homeostasis and the formation of tumors.
Collapse
Affiliation(s)
- Moritz Sander
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Teresa Eichenlaub
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
STAT, Wingless, and Nurf-38 determine the accuracy of regeneration after radiation damage in Drosophila. PLoS Genet 2017; 13:e1007055. [PMID: 29028797 PMCID: PMC5656321 DOI: 10.1371/journal.pgen.1007055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/25/2017] [Accepted: 10/04/2017] [Indexed: 01/01/2023] Open
Abstract
We report here a study of regeneration in Drosophila larval wing imaginal discs after damage by ionizing radiation. We detected faithful regeneration that restored a wing disc and abnormal regeneration that produced an extra wing disc. We describe a sequence of changes in cell number, location and fate that occur to produce an ectopic disc. We identified a group of cells that not only participate in ectopic disc formation but also recruit others to do so. STAT92E (Drosophila STAT3/5) and Nurf-38, which encodes a member of the Nucleosome Remodeling Factor complex, oppose each other in these cells to modulate the frequency of ectopic disc growth. The picture that emerges is one in which activities like STAT increase after radiation damage and fulfill essential roles in rebuilding the tissue. But such activities must be kept in check so that one and only one wing disc is regenerated. Accuracy in regeneration ensures that the original structures are restored, no more and no less. Prior studies in the wing primordia of Drosophila melanogaster larvae that have been damaged by high energy radiation show that regeneration occurs to restore the original structure. We report here that, in the same experimental system, abnormal regeneration can also occur to produce extra wing structures. We describe a series of cell rearrangements and fate changes that underlie abnormal regeneration, and identify genes responsible for these events. Modulation of such genes have the potential to mitigate abnormal regeneration that occurs after radiation damage to produce such side effects as ulcers and fibrosis.
Collapse
|
30
|
Martín R, Pinal N, Morata G. Distinct regenerative potential of trunk and appendages of Drosophila mediated by JNK signalling. Development 2017; 144:3946-3956. [PMID: 28935711 DOI: 10.1242/dev.155507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/11/2017] [Indexed: 12/31/2022]
Abstract
The Drosophila body comprises a central part, the trunk, and outgrowths of the trunk, the appendages. Much is known about appendage regeneration, but little about the trunk. As the wing imaginal disc contains a trunk component, the notum, and a wing appendage, we have investigated the response to ablation of these two components. We find that, in contrast with the strong regenerative response of the wing, the notum does not regenerate. Nevertheless, the elimination of the wing primordium elicits a proliferative response of notum cells, but they do not regenerate wing; they form a notum duplicate. Conversely, the wing cells cannot regenerate an ablated notum; they overproliferate and generate a hinge overgrowth. These results suggest that trunk and appendages cannot be reprogrammed to generate each other. Our experiments demonstrate that the proliferative response is mediated by JNK signalling from dying cells, but JNK functions differently in the trunk and the appendages, which may explain their distinct regenerative potential.
Collapse
Affiliation(s)
- Raquel Martín
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Noelia Pinal
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ginés Morata
- Centro de Biología Molecular CSIC-UAM, Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
31
|
Deshmukh R, Baral S, Gandhimathi A, Kuwalekar M, Kunte K. Mimicry in butterflies: co-option and a bag of magnificent developmental genetic tricks. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 7. [PMID: 28913870 DOI: 10.1002/wdev.291] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 07/04/2017] [Accepted: 07/20/2017] [Indexed: 01/05/2023]
Abstract
Butterfly wing patterns are key adaptations that are controlled by remarkable developmental and genetic mechanisms that facilitate rapid evolutionary change. With swift advancements in the fields of genomics and genetic manipulations, identifying the regulators of wing development and mimetic wing patterns has become feasible even in nonmodel organisms such as butterflies. Recent mapping and gene expression studies have identified single switch loci of major effects such as transcription factors and supergenes as the main drivers of adaptive evolution of mimetic and polymorphic butterfly wing patterns. We highlight several of these examples, with emphasis on doublesex, optix, WntA and other dynamic, yet essential, master regulators that control critical color variation and sex-specific traits. Co-option emerges as a predominant theme, where typically embryonic and other early-stage developmental genes and networks have been rewired to regulate polymorphic and sex-limited mimetic wing patterns in iconic butterfly adaptations. Drawing comparisons from our knowledge of wing development in Drosophila, we illustrate the functional space of genes that have been recruited to regulate butterfly wing patterns. We also propose a developmental pathway that potentially results in dorsoventral mismatch in butterfly wing patterns. Such dorsoventrally mismatched color patterns modulate signal components of butterfly wings that are used in intra- and inter-specific communication. Recent advances-fuelled by RNAi-mediated knockdowns and CRISPR/Cas9-based genomic edits-in the developmental genetics of butterfly wing patterns, and the underlying biological diversity and complexity of wing coloration, are pushing butterflies as an emerging model system in ecological genetics and evolutionary developmental biology. WIREs Dev Biol 2018, 7:e291. doi: 10.1002/wdev.291 This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Comparative Development and Evolution > Regulation of Organ Diversity Comparative Development and Evolution > Evolutionary Novelties.
Collapse
Affiliation(s)
| | - Saurav Baral
- National Centre for Biological Sciences, Bengaluru, India
| | - A Gandhimathi
- National Centre for Biological Sciences, Bengaluru, India
| | | | | |
Collapse
|
32
|
Abstract
While several large-scale resources are available for in vivo loss-of-function studies in Drosophila, an analogous resource for overexpressing genes from their endogenous loci does not exist. We describe a strategy for generating such a resource using Cas9 transcriptional activators (CRISPRa). First, we compare a panel of CRISPRa approaches and demonstrate that, for in vivo studies, dCas9-VPR is the most optimal activator. Next, we demonstrate that this approach is scalable and has a high success rate, as >75% of the lines tested activate their target gene. We show that CRISPRa leads to physiologically relevant levels of target gene expression capable of generating strong gain-of-function (GOF) phenotypes in multiple tissues and thus serves as a useful platform for genetic screening. Based on the success of this CRISRPa approach, we are generating a genome-wide collection of flies expressing single-guide RNAs (sgRNAs) for CRISPRa. We also present a collection of more than 30 Gal4 > UAS:dCas9-VPR lines to aid in using these sgRNA lines for GOF studies in vivo.
Collapse
|
33
|
Song S, Herranz H, Cohen SM. The chromatin remodeling BAP complex limits tumor-promoting activity of the Hippo pathway effector Yki to prevent neoplastic transformation in Drosophila epithelia. Dis Model Mech 2017; 10:1201-1209. [PMID: 28754838 PMCID: PMC5665456 DOI: 10.1242/dmm.030122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/21/2017] [Indexed: 12/26/2022] Open
Abstract
Switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complexes are mutated in many human cancers. In this article, we make use of a Drosophila genetic model for epithelial tumor formation to explore the tumor suppressive role of SWI/SNF complex proteins. Members of the BAP complex exhibit tumor suppressor activity in tissue overexpressing the Yorkie (Yki) proto-oncogene, but not in tissue overexpressing epidermal growth factor receptor (EGFR). The Brahma-associated protein (BAP) complex has been reported to serve as a Yki-binding cofactor to support Yki target expression. However, we observed that depletion of BAP leads to ectopic expression of Yki targets both autonomously and non-autonomously, suggesting additional indirect effects. We provide evidence that BAP complex depletion causes upregulation of the Wingless (Wg) and Decapentaplegic (Dpp) morphogens to promote tumor formation in cooperation with Yki.
Collapse
Affiliation(s)
- Shilin Song
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200N, Denmark
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200N, Denmark
| | - Stephen M Cohen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200N, Denmark
| |
Collapse
|
34
|
Barrio L, Milán M. Boundary Dpp promotes growth of medial and lateral regions of the Drosophila wing. eLife 2017; 6:22013. [PMID: 28675372 PMCID: PMC5560857 DOI: 10.7554/elife.22013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 06/04/2017] [Indexed: 11/14/2022] Open
Abstract
The gradient of Decapentaplegic (Dpp) in the Drosophila wing has served as a paradigm to characterize the role of morphogens in regulating patterning. However, the role of this gradient in regulating tissue size is a topic of intense debate as proliferative growth is homogenous. Here, we combined the Gal4/UAS system and a temperature-sensitive Gal80 molecule to induce RNAi-mediated depletion of dpp and characterise the spatial and temporal requirement of Dpp in promoting growth. We show that Dpp emanating from the AP compartment boundary is required throughout development to promote growth by regulating cell proliferation and tissue size. Dpp regulates growth and proliferation rates equally in central and lateral regions of the developing wing appendage and reduced levels of Dpp affects similarly the width and length of the resulting wing. We also present evidence supporting the proposal that graded activity of Dpp is not an absolute requirement for wing growth. DOI:http://dx.doi.org/10.7554/eLife.22013.001 From the wings of a butterfly to the fingers of a human hand, living tissues often have complex and intricate patterns. Developmental biologists have long been fascinated by the signals – called morphogens – that guide how these kinds of pattern develop. Morphogens are substances that are produced by groups of cells and spread to the rest of the tissue to form a gradient. Depending on where they sit along this gradient, cells in the tissue activate different sets of genes, and the resulting pattern of gene activity ultimately defines the position of the different parts of the tissue. Decades worth of studies into how limbs develop in animals from mice to fruit flies have revealed common principles of morphogen gradients that regulate the development of tissue patterns. Morphogens have been shown to help regulate the growth of tissues in a number of different animals as well. However, how the morphogens regulate tissue size and what role their gradients play in this process remain topics of intense debate in the field of developmental biology. In the developing wing of a fruit fly, a morphogen called Dpp is expressed in a thin stripe located in the center and spreads to the rest of the tissue to form a gradient. Barrio and Milán have now characterized where and when the Dpp morphogen must be produced to regulate both the final size of the fly’s wing and the number of cells the wing eventually contains. The experiments involved preventing the production of Dpp in the developing wing in specific cells and at specific stages of development. This approach confirmed that Dpp must be produced in the central stripe for the wing to grow. Matsuda and Affolter and, independently, Bosch, Ziukaite, Alexandre et al. report the same findings in two related studies. Moreover, Barrio and Milán and Bosch et al. also conclude that the gradient of Dpp throughout the wing is not required for growth. Further work will be needed to explain how the Dpp signal regulates the growth of the wing. The answer to this question will contribute to a better understanding of the role of morphogens in regulating the size of human organs and how a failure to do so might cause developmental disorders. DOI:http://dx.doi.org/10.7554/eLife.22013.002
Collapse
Affiliation(s)
- Lara Barrio
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain.,The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| |
Collapse
|
35
|
Cap-n-Collar Promotes Tissue Regeneration by Regulating ROS and JNK Signaling in the Drosophila melanogaster Wing Imaginal Disc. Genetics 2017; 206:1505-1520. [PMID: 28512185 DOI: 10.1534/genetics.116.196832] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/10/2017] [Indexed: 01/03/2023] Open
Abstract
Regeneration is a complex process that requires an organism to recognize and repair tissue damage, as well as grow and pattern new tissue. Here, we describe a genetic screen to identify novel regulators of regeneration. We ablated the Drosophila melanogaster larval wing primordium by inducing apoptosis in a spatially and temporally controlled manner and allowed the tissue to regenerate and repattern. To identify genes that regulate regeneration, we carried out a dominant-modifier screen by assessing the amount and quality of regeneration in adult wings heterozygous for isogenic deficiencies. We have identified 31 regions on the right arm of the third chromosome that modify the regenerative response. Interestingly, we observed several distinct phenotypes: mutants that regenerated poorly, mutants that regenerated faster or better than wild-type, and mutants that regenerated imperfectly and had patterning defects. We mapped one deficiency region to cap-n-collar (cnc), the Drosophila Nrf2 ortholog, which is required for regeneration. Cnc regulates reactive oxygen species levels in the regenerating epithelium, and affects c-Jun N-terminal protein kinase (JNK) signaling, growth, debris localization, and pupariation timing. Here, we present the results of our screen and propose a model wherein Cnc regulates regeneration by maintaining an optimal level of reactive oxygen species to promote JNK signaling.
Collapse
|
36
|
JAK/STAT controls organ size and fate specification by regulating morphogen production and signalling. Nat Commun 2017; 8:13815. [PMID: 28045022 PMCID: PMC5216089 DOI: 10.1038/ncomms13815] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2017] [Indexed: 01/20/2023] Open
Abstract
A stable pool of morphogen-producing cells is critical for the development of any organ or tissue. Here we present evidence that JAK/STAT signalling in the Drosophila wing promotes the cycling and survival of Hedgehog-producing cells, thereby allowing the stable localization of the nearby BMP/Dpp-organizing centre in the developing wing appendage. We identify the inhibitor of apoptosis dIAP1 and Cyclin A as two critical genes regulated by JAK/STAT and contributing to the growth of the Hedgehog-expressing cell population. We also unravel an early role of JAK/STAT in guaranteeing Wingless-mediated appendage specification, and a later one in restricting the Dpp-organizing activity to the appendage itself. These results unveil a fundamental role of the conserved JAK/STAT pathway in limb specification and growth by regulating morphogen production and signalling, and a function of pro-survival cues and mitogenic signals in the regulation of the pool of morphogen-producing cells in a developing organ.
Collapse
|
37
|
Pflugfelder G, Eichinger F, Shen J. T-Box Genes in Drosophila Limb Development. Curr Top Dev Biol 2017; 122:313-354. [DOI: 10.1016/bs.ctdb.2016.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
Ribeiro AL, Silva RD, Foyn H, Tiago MN, Rathore OS, Arnesen T, Martinho RG. Naa50/San-dependent N-terminal acetylation of Scc1 is potentially important for sister chromatid cohesion. Sci Rep 2016; 6:39118. [PMID: 27996020 PMCID: PMC5171793 DOI: 10.1038/srep39118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/17/2016] [Indexed: 12/17/2022] Open
Abstract
The gene separation anxiety (san) encodes Naa50/San, a N-terminal acetyltransferase required for chromosome segregation during mitosis. Although highly conserved among higher eukaryotes, the mitotic function of this enzyme is still poorly understood. Naa50/San was originally proposed to be required for centromeric sister chromatid cohesion in Drosophila and human cells, yet, more recently, it was also suggested to be a negative regulator of microtubule polymerization through internal acetylation of beta Tubulin. We used genetic and biochemical approaches to clarify the function of Naa50/San during development. Our work suggests that Naa50/San is required during tissue proliferation for the correct interaction between the cohesin subunits Scc1 and Smc3. Our results also suggest a working model where Naa50/San N-terminally acetylates the nascent Scc1 polypeptide, and that this co-translational modification is subsequently required for the establishment and/or maintenance of sister chromatid cohesion.
Collapse
Affiliation(s)
- Ana Luisa Ribeiro
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.,Center for Biomedical Research (CBMR), University of Algarve, 8005-139 Faro, Portugal
| | - Rui D Silva
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.,Center for Biomedical Research (CBMR), University of Algarve, 8005-139 Faro, Portugal
| | - Håvard Foyn
- Department of Molecular Biology, University of Bergen, N-5020 Bergen, Norway
| | - Margarida N Tiago
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.,Center for Biomedical Research (CBMR), University of Algarve, 8005-139 Faro, Portugal
| | - Om Singh Rathore
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.,Center for Biomedical Research (CBMR), University of Algarve, 8005-139 Faro, Portugal
| | - Thomas Arnesen
- Department of Molecular Biology, University of Bergen, N-5020 Bergen, Norway.,Department of Surgery, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Rui Gonçalo Martinho
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.,Center for Biomedical Research (CBMR), University of Algarve, 8005-139 Faro, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
39
|
Fabian B, Schneeberg K, Beutel RG. Comparative thoracic anatomy of the wild type and wingless (wg 1cn 1) mutant of Drosophila melanogaster (Diptera). ARTHROPOD STRUCTURE & DEVELOPMENT 2016; 45:611-636. [PMID: 27720953 DOI: 10.1016/j.asd.2016.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/29/2016] [Accepted: 10/04/2016] [Indexed: 06/06/2023]
Abstract
Genetically modified organisms are crucial for our understanding of gene regulatory networks, physiological processes and ontogeny. With modern molecular genetic techniques allowing the rapid generation of different Drosophila melanogaster mutants, efficient in-depth morphological investigations become an important issue. Anatomical studies can elucidate the role of certain genes in developmental processes and point out which parts of gene regulatory networks are involved in evolutionary changes of morphological structures. The wingless mutation wg1 of D. melanogaster was discovered more than 40 years ago. While early studies addressed the external phenotype of these mutants, the documentation of the internal organization was largely restricted to the prominent indirect flight muscles. We used SEM micrographs, histological serial sections, μ-computed tomography, CLSM and 3D reconstructions to study and document the thoracic skeletomuscular system of the wild type and mutant. A recently introduced nomenclature for the musculature of neopteran insects was applied to facilitate comparisons with closely or more distantly related taxa. The mutation is phenotypically mainly characterized by the absence of one or both wings and halteres. The wing is partly or entirely replaced by duplications of mesonotal structures, whereas the haltere and its associated muscles are completely absent on body sides showing the reduction. Both the direct and indirect mesothoracic flight muscles are affected by loss and reorientation of bundles or fibers. Our observations lead to the conclusion that the wingless mutation causes a homeotic transformation in the imaginal discs of wings and halteres with a direct effect on the development of skeletal structures and an indirect effect on the associated muscular system.
Collapse
Affiliation(s)
- Benjamin Fabian
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, FSU Jena, Erbertstr. 1, 07743 Jena, Germany.
| | - Katharina Schneeberg
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, FSU Jena, Erbertstr. 1, 07743 Jena, Germany.
| | - Rolf Georg Beutel
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, FSU Jena, Erbertstr. 1, 07743 Jena, Germany.
| |
Collapse
|
40
|
Benham-Pyle BW, Sim JY, Hart KC, Pruitt BL, Nelson WJ. Increasing β-catenin/Wnt3A activity levels drive mechanical strain-induced cell cycle progression through mitosis. eLife 2016; 5. [PMID: 27782880 PMCID: PMC5104517 DOI: 10.7554/elife.19799] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/25/2016] [Indexed: 11/13/2022] Open
Abstract
Mechanical force and Wnt signaling activate β-catenin-mediated transcription to promote proliferation and tissue expansion. However, it is unknown whether mechanical force and Wnt signaling act independently or synergize to activate β-catenin signaling and cell division. We show that mechanical strain induced Src-dependent phosphorylation of Y654 β-catenin and increased β-catenin-mediated transcription in mammalian MDCK epithelial cells. Under these conditions, cells accumulated in S/G2 (independent of DNA damage) but did not divide. Activating β-catenin through Casein Kinase I inhibition or Wnt3A addition increased β-catenin-mediated transcription and strain-induced accumulation of cells in S/G2. Significantly, only the combination of mechanical strain and Wnt/β-catenin activation triggered cells in S/G2 to divide. These results indicate that strain-induced Src phosphorylation of β-catenin and Wnt-dependent β-catenin stabilization synergize to increase β-catenin-mediated transcription to levels required for mitosis. Thus, local Wnt signaling may fine-tune the effects of global mechanical strain to restrict cell divisions during tissue development and homeostasis. DOI:http://dx.doi.org/10.7554/eLife.19799.001 Tissues and organs can both produce and respond to physical forces. For example, the lungs expand and contract; the heart pumps blood; and bones and muscles grow or shrink depending on how much they are used. These responses are possible because cells contain proteins that can respond to physical forces. One of the best studied of these is a protein called β-catenin, which increases the activity of genes that trigger cells to divide to promote the expansion of tissues. β-catenin is over-active in many types of cancer cells where it contributes to tumor growth. In addition to being switched on by mechanical force, β-catenin is also activated when cells detect a signal molecule called Wnt. Cells cycle through a series of stages known as the cell cycle to ensure that they only divide when they are fully prepared to do so. Benham-Pyle et al. investigated if physical force and Wnt activate β-catenin in the same way or if they have different effects on cell division. The experiments were conducted on dog kidney cells that had left the cell cycle and had therefore temporarily stopped dividing. Physical forces, such as stretching, resulted in β-catenin being modified by an enzyme called SRC kinase, which allowed the cells to re-enter the cell cycle. On the other hand, Wnt stabilized β-catenin and temporarily increased the number of cell divisions. When mechanical stretch and Wnt signaling were combined, the cells were more likely to re-enter the cell cycle and divide compared to either stimulus alone. These data suggest that physical force and Wnt signaling affect β-catenin differently and that they can therefore have a greater effect on cell or tissue growth when they act together than on their own. The findings of Benham-Pyle et al. show that β-catenin is not simply switched on or off, but can have different levels of activity depending on the input the cells are receiving. Future experiments will test whether these mechanisms also exist in three-dimensional tissues, which will help us understand how organs develop. DOI:http://dx.doi.org/10.7554/eLife.19799.002
Collapse
Affiliation(s)
| | - Joo Yong Sim
- Department of Mechanical Engineering, Stanford University, Stanford, United States
| | - Kevin C Hart
- Department of Biology, Stanford University, Stanford, United States
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, United States.,Stanford Cardiovascular Institute, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - William James Nelson
- Program in Cancer Biology, Stanford University, Stanford, United States.,Department of Biology, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| |
Collapse
|
41
|
Simon E, Faucheux C, Zider A, Thézé N, Thiébaud P. From vestigial to vestigial-like: the Drosophila gene that has taken wing. Dev Genes Evol 2016; 226:297-315. [PMID: 27116603 DOI: 10.1007/s00427-016-0546-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/10/2016] [Indexed: 12/16/2022]
Abstract
The members of the vestigial-like gene family have been identified as homologs of the Drosophila vestigial, which is essential to wing formation. All members of the family are characterized by the presence of the TONDU domain, a highly conserved sequence that mediates their interaction with the transcription factors of the TEAD family. Mammals possess four vestigial-like genes that can be subdivided into two classes, depending on the number of Tondu domains present. While vestigial proteins have been studied in great depth in Drosophila, we still have sketchy knowledge of the functions of vestigial-like proteins in vertebrates. Recent studies have unveiled unexpected functions for some of these members and reveal the role they play in the Hippo pathway. Here, we present the current knowledge about vestigial-like family gene members and their functions, together with their identification in different taxa.
Collapse
Affiliation(s)
- Emilie Simon
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Corinne Faucheux
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Alain Zider
- Univ Paris Diderot, Sorbonne Paris Cité, IJM, UMR 7592 CNRS, F-75205, Paris, France
| | - Nadine Thézé
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France
| | - Pierre Thiébaud
- Univ. Bordeaux, BMGIC, U1035, F-33000, Bordeaux, France.
- INSERM, BMGIC, U1035, F-33000, Bordeaux, France.
- Univ. Bordeaux, INSERM U1035, 146 rue Léo Saignat, 33076, Bordeaux CEDEX, France.
| |
Collapse
|
42
|
Lin S, Ewen-Campen B, Ni X, Housden BE, Perrimon N. In Vivo Transcriptional Activation Using CRISPR/Cas9 in Drosophila. Genetics 2015; 201:433-42. [PMID: 26245833 PMCID: PMC4596659 DOI: 10.1534/genetics.115.181065] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/04/2015] [Indexed: 12/30/2022] Open
Abstract
A number of approaches for Cas9-mediated transcriptional activation have recently been developed, allowing target genes to be overexpressed from their endogenous genomic loci. However, these approaches have thus far been limited to cell culture, and this technique has not been demonstrated in vivo in any animal. The technique involving the fewest separate components, and therefore the most amenable to in vivo applications, is the dCas9-VPR system, where a nuclease-dead Cas9 is fused to a highly active chimeric activator domain. In this study, we characterize the dCas9-VPR system in Drosophila cells and in vivo. We show that this system can be used in cell culture to upregulate a range of target genes, singly and in multiplex, and that a single guide RNA upstream of the transcription start site can activate high levels of target transcription. We observe marked heterogeneity in guide RNA efficacy for any given gene, and we confirm that transcription is inhibited by guide RNAs binding downstream of the transcription start site. To demonstrate one application of this technique in cells, we used dCas9-VPR to identify target genes for Twist and Snail, two highly conserved transcription factors that cooperate during Drosophila mesoderm development. In addition, we simultaneously activated both Twist and Snail to identify synergistic responses to this physiologically relevant combination. Finally, we show that dCas9-VPR can activate target genes and cause dominant phenotypes in vivo, providing the first demonstration of dCas9 activation in a multicellular animal. Transcriptional activation using dCas9-VPR thus offers a simple and broadly applicable technique for a variety of overexpression studies.
Collapse
Affiliation(s)
- Shuailiang Lin
- Tsinghua-Peking-National Institute of Biological Sciences Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China Department of Genetics
| | | | | | | | - Norbert Perrimon
- Department of Genetics Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
43
|
cis-regulatory analysis of the Drosophila pdm locus reveals a diversity of neural enhancers. BMC Genomics 2015; 16:700. [PMID: 26377945 PMCID: PMC4574355 DOI: 10.1186/s12864-015-1897-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/02/2015] [Indexed: 11/15/2022] Open
Abstract
Background One of the major challenges in developmental biology is to understand the regulatory events that generate neuronal diversity. During Drosophila embryonic neural lineage development, cellular temporal identity is established in part by a transcription factor (TF) regulatory network that mediates a cascade of cellular identity decisions. Two of the regulators essential to this network are the POU-domain TFs Nubbin and Pdm-2, encoded by adjacent genes collectively known as pdm. The focus of this study is the discovery and characterization of cis-regulatory DNA that governs their expression. Results Phylogenetic footprinting analysis of a 125 kb genomic region that spans the pdm locus identified 116 conserved sequence clusters. To determine which of these regions function as cis-regulatory enhancers that regulate the dynamics of pdm gene expression, we tested each for in vivo enhancer activity during embryonic development and postembryonic neurogenesis. Our screen revealed 77 unique enhancers positioned throughout the noncoding region of the pdm locus. Many of these activated neural-specific gene expression during different developmental stages and many drove expression in overlapping patterns. Sequence comparisons of functionally related enhancers that activate overlapping expression patterns revealed that they share conserved elements that can be predictive of enhancer behavior. To facilitate data accessibility, the results of our analysis are catalogued in cisPatterns, an online database of the structure and function of these and other Drosophila enhancers. Conclusions These studies reveal a diversity of modular enhancers that most likely regulate pdm gene expression during embryonic and adult development, highlighting a high level of temporal and spatial expression specificity. In addition, we discovered clusters of functionally related enhancers throughout the pdm locus. A subset of these enhancers share conserved elements including sequences that correspond to known TF DNA binding sites. Although comparative analysis of the nubbin and pdm-2 encoding sequences indicate that these two genes most likely arose from a duplication event, we found only partial evidence of sequence duplication between their enhancers, suggesting that after the putative duplication their cis-regulatory DNA diverged at a higher rate than their coding sequences. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1897-2) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Yang Y, Wilson MJ. Lhx9 gene expression during early limb development in mice requires the FGF signalling pathway. Gene Expr Patterns 2015. [DOI: 10.1016/j.gep.2015.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Dally Proteoglycan Mediates the Autonomous and Nonautonomous Effects on Tissue Growth Caused by Activation of the PI3K and TOR Pathways. PLoS Biol 2015; 13:e1002239. [PMID: 26313758 PMCID: PMC4551486 DOI: 10.1371/journal.pbio.1002239] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 07/31/2015] [Indexed: 11/19/2022] Open
Abstract
How cells acquiring mutations in tumor suppressor genes outcompete neighboring wild-type cells is poorly understood. The phosphatidylinositol 3-kinase (PI3K)–phosphatase with tensin homology (PTEN) and tuberous sclerosis complex (TSC)-target of rapamycin (TOR) pathways are frequently activated in human cancer, and this activation is often causative of tumorigenesis. We utilized the Gal4-UAS system in Drosophila imaginal primordia, highly proliferative and growing tissues, to analyze the impact of restricted activation of these pathways on neighboring wild-type cell populations. Activation of these pathways leads to an autonomous induction of tissue overgrowth and to a remarkable nonautonomous reduction in growth and proliferation rates of adjacent cell populations. This nonautonomous response occurs independently of where these pathways are activated, is functional all throughout development, takes place across compartments, and is distinct from cell competition. The observed autonomous and nonautonomous effects on tissue growth rely on the up-regulation of the proteoglycan Dally, a major element involved in modulating the spreading, stability, and activity of the growth promoting Decapentaplegic (Dpp)/transforming growth factor β(TGF-β) signaling molecule. Our findings indicate that a reduction in the amount of available growth factors contributes to the outcompetition of wild-type cells by overgrowing cell populations. During normal development, the PI3K/PTEN and TSC/TOR pathways play a major role in sensing nutrient availability and modulating the final size of any developing organ. We present evidence that Dally also contributes to integrating nutrient sensing and organ scaling, the fitting of pattern to size. The loss of tumor suppressor genes induces a nonautonomous reduction of growth and proliferation rates in adjacent cell populations by competing for available growth factors; the proteoglycan Dally helps to mediate this effect. The final size of a developing organ is finely modulated by nutrient conditions through the activity of nutrient sensing pathways, and deregulation of these pathways is often causative of tumorigenesis. Besides the well-known roles of these pathways in inducing tissue and cell growth, here we identify a nonautonomous effect of activation of these pathways on growth and proliferation rates and on the final size of neighboring cell populations. We reveal that the observed autonomous and nonautonomous effects on tissue growth and proliferation rates rely on the up-regulation of the proteoglycan Dally, a major factor involved in modulating the spreading, stability, and activity of the growth promoting Decapentaplegic (Dpp)/transforming growth factor β(TGF-β) signaling molecule. Our data indicate that a reduction in the amount of available growth factors contributes to the outcompetition of wild-type cells by overgrowing cell populations. Whereas nutrient-sensing pathways modulate the final size of the adult structure according to nutrient availability to the feeding animal, Dpp plays an organ-intrinsic role in the coordination of growth and patterning. We identify the proteoglycan Dally as the rate-limiting factor that contributes to the tissue-autonomous and nonautonomous effects on growth caused by targeted activation of the nutrient-sensing pathways. Thus, our results unravel a role of Dally as a molecular bridge between the organ-intrinsic and organ-extrinsic mechanisms that regulate organ size.
Collapse
|
46
|
Huang H, Kornberg TB. Myoblast cytonemes mediate Wg signaling from the wing imaginal disc and Delta-Notch signaling to the air sac primordium. eLife 2015; 4:e06114. [PMID: 25951303 PMCID: PMC4423120 DOI: 10.7554/elife.06114] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/16/2015] [Indexed: 12/17/2022] Open
Abstract
The flight muscles, dorsal air sacs, wing blades, and thoracic cuticle of the Drosophila adult function in concert, and their progenitor cells develop together in the wing imaginal disc. The wing disc orchestrates dorsal air sac development by producing decapentaplegic and fibroblast growth factor that travel via specific cytonemes in order to signal to the air sac primordium (ASP). Here, we report that cytonemes also link flight muscle progenitors (myoblasts) to disc cells and to the ASP, enabling myoblasts to relay signaling between the disc and the ASP. Frizzled (Fz)-containing myoblast cytonemes take up Wingless (Wg) from the disc, and Delta (Dl)-containing myoblast cytonemes contribute to Notch activation in the ASP. Wg signaling negatively regulates Dl expression in the myoblasts. These results reveal an essential role for cytonemes in Wg and Notch signaling and for a signal relay system in the myoblasts.
Collapse
Affiliation(s)
- Hai Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Thomas B Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
47
|
Ruiz-Romero M, Blanco E, Paricio N, Serras F, Corominas M. Cabut/dTIEG associates with the transcription factor Yorkie for growth control. EMBO Rep 2015; 16:362-369. [PMID: 25572844 PMCID: PMC4364875 DOI: 10.15252/embr.201439193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 11/24/2014] [Accepted: 12/08/2014] [Indexed: 11/21/2022] Open
Abstract
The Drosophila transcription factor Cabut/dTIEG (Cbt) is a growth regulator, whose expression is modulated by different stimuli. Here, we determine Cbt association with chromatin and identify Yorkie (Yki), the transcriptional co-activator of the Hippo (Hpo) pathway as its partner. Cbt and Yki co-localize on common gene promoters, and the expression of target genes varies according to changes in Cbt levels. Down-regulation of Cbt suppresses the overgrowth phenotypes caused by mutations in expanded (ex) and yki overexpression, whereas its up-regulation promotes cell proliferation. Our results imply that Cbt is a novel partner of Yki that is required as a transcriptional co-activator in growth control.
Collapse
Affiliation(s)
- Marina Ruiz-Romero
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina (IBUB) de la Universitat de Barcelona, Barcelona, Spain
| | - Enrique Blanco
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina (IBUB) de la Universitat de Barcelona, Barcelona, Spain Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad de Ciencias Biológicas, Universidad de Valencia, Valencia, Spain
| | - Florenci Serras
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina (IBUB) de la Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Corominas
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina (IBUB) de la Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Caine C, Kasherov P, Silber J, Lalouette A. Mef2 interacts with the Notch pathway during adult muscle development in Drosophila melanogaster. PLoS One 2014; 9:e108149. [PMID: 25247309 PMCID: PMC4172597 DOI: 10.1371/journal.pone.0108149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 08/03/2014] [Indexed: 12/22/2022] Open
Abstract
Myogenesis of indirect flight muscles (IFMs) in Drosophila melanogaster follows a well-defined cellular developmental scheme. During embryogenesis, a set of cells, the Adult Muscle Precursors (AMPs), are specified. These cells will become proliferating myoblasts during the larval stages which will then give rise to the adult IFMs. Although the cellular aspect of this developmental process is well studied, the molecular biology behind the different stages is still under investigation. In particular, the interactions required during the transition from proliferating myoblasts to differentiated myoblasts ready to fuse to the muscle fiber. It has been previously shown that the Notch pathway is active in proliferating myoblasts, and that this pathway is inhibited in developing muscle fibers. Furthermore, the Myocyte Enhancing Factor 2 (Mef2), Vestigial (Vg) and Scalloped (Sd) transcription factors are necessary for IFM development and that Vg is required for Notch pathway repression in differentiating fibers. Here we examine the interactions between Notch and Mef2 and mechanisms by which the Notch pathway is inhibited during differentiation. We show that Mef2 is capable of inhibiting the Notch pathway in non myogenic cells. A previous screen for Mef2 potential targets identified Delta a component of the Notch pathway. Dl is expressed in Mef2 and Sd-positive developing fibers. Our results show that Mef2 and possibly Sd regulate a Dl enhancer specifically expressed in the developing IFMs and that Mef2 is required for Dl expression in developing IFMs.
Collapse
Affiliation(s)
- Charlotte Caine
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Petar Kasherov
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Joël Silber
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Alexis Lalouette
- Institut Jacques Monod, CNRS, UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
49
|
The role of Bro1- domain-containing protein Myopic in endosomal trafficking of Wnt/Wingless. Dev Biol 2014; 392:93-107. [DOI: 10.1016/j.ydbio.2014.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/24/2014] [Accepted: 04/25/2014] [Indexed: 11/19/2022]
|
50
|
Oliveira MM, Shingleton AW, Mirth CK. Coordination of wing and whole-body development at developmental milestones ensures robustness against environmental and physiological perturbations. PLoS Genet 2014; 10:e1004408. [PMID: 24945255 PMCID: PMC4063698 DOI: 10.1371/journal.pgen.1004408] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/14/2014] [Indexed: 11/18/2022] Open
Abstract
Development produces correctly patterned tissues under a wide range of conditions that alter the rate of development in the whole body. We propose two hypotheses through which tissue patterning could be coordinated with whole-body development to generate this robustness. Our first hypothesis states that tissue patterning is tightly coordinated with whole-body development over time. The second hypothesis is that tissue patterning aligns at developmental milestones. To distinguish between our two hypotheses, we developed a staging scheme for the wing imaginal discs of Drosophila larvae using the expression of canonical patterning genes, linking our scheme to three whole-body developmental events: moulting, larval wandering and pupariation. We used our scheme to explore how the progression of pattern changes when developmental time is altered either by changing temperature or by altering the timing of hormone synthesis that drives developmental progression. We found the expression pattern in the wing disc always aligned at moulting and pupariation, indicating that these key developmental events represent milestones. Between these milestones, the progression of pattern showed greater variability in response to changes in temperature and alterations in physiology. Furthermore, our data showed that discs from wandering larvae showed greater variability in patterning stage. Thus for wing disc patterning, wandering does not appear to be a developmental milestone. Our findings reveal that tissue patterning remains robust against environmental and physiological perturbations by aligning at developmental milestones. Furthermore, our work provides an important glimpse into how the development of individual tissues is coordinated with the body as a whole.
Collapse
Affiliation(s)
- Marisa M. Oliveira
- Development, Evolution and the Environment Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Alexander W. Shingleton
- Dept. of Zoology, Michigan State University, East Lansing, Michigan, United States of America
- Dept. of Biology, Lake Forest College, Lake Forest, Illinois, United States of America
| | - Christen K. Mirth
- Development, Evolution and the Environment Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
- * E-mail:
| |
Collapse
|