1
|
Dong Y, Huang L, Liu L. Comparative analysis of testicular fusion in Spodoptera litura (cutworm) and Bombyx mori (silkworm): Histological and transcriptomic insights. Gen Comp Endocrinol 2024; 356:114562. [PMID: 38848820 DOI: 10.1016/j.ygcen.2024.114562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Spodoptera litura commonly known as the cutworm, is among the most destructive lepidopteran pests affecting over 120 plants species. The powerful destructive nature of this lepidopteran is attributable to its high reproductive capacity. The testicular fusion that occurs during metamorphosis from larvae to pupa in S.litura positively influences the reproductive success of the offspring. In contrast, Bombyx mori, the silkworm, retains separate testes throughout its life and does not undergo this fusion process. Microscopic examination reveals that during testicular fusion in S.litura, the peritoneal sheath becomes thinner and more translucent, whereas in B.mori, the analogous region thickens. The outer basement membrane in S.litura exhibits fractures, discontinuity, and uneven thickness accompanied by a significant presence of cellular secretions, large cell size, increased vesicles, liquid droplets, and a proliferation of rough endoplasmic reticulum and mitochondria. In contrast, the testicular peritoneal sheath of B.mori at comparable developmental stage exhibits minimal change. Comparative transcriptomic analysis of the testicular peritoneal sheath reveals a substantial difference in gene expression between the two species. The disparity in differential expressed genes (DEGs) is linked to an enrichment of numerous transcription factors, intracellular signaling pathways involving Ca2+ and GTPase, as well as intracellular protein transport and signaling pathways. Meanwhile, structural proteins including actin, chitin-binding proteins, membrane protein fractions, cell adhesion, extracellular matrix proteins are predominantly identified. Moreover, the study highlights the enrichment of endopeptidases, serine proteases, proteolytic enzymes and matrix metalloproteins, which may play a role in the degradation of the outer membrane. Five transcription factors-Slforkhead, Slproline, Slcyclic, Slsilk, and SlD-ETS were identified, and their expression pattern were confirmed by qRT-PCR. they are candidates for participating in the regulation of testicular fusion. Our findings underscore significant morphological and trancriptomic variation in the testicular peritoneal sheath of S.litura compared to the silkworm, with substantial changes at the transcriptomic level coinciding with testicular fusion. The research provides valuable clues for understanding the complex mechanisms underlying this unique phenomenon in insects.
Collapse
Affiliation(s)
- Yaqun Dong
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lihua Huang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lin Liu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
2
|
Karkali K, Pastor-Pareja JC, Martin-Blanco E. JNK signaling and integrins cooperate to maintain cell adhesion during epithelial fusion in Drosophila. Front Cell Dev Biol 2024; 11:1034484. [PMID: 38264353 PMCID: PMC10803605 DOI: 10.3389/fcell.2023.1034484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
The fusion of epithelial sheets is an essential and conserved morphogenetic event that requires the maintenance of tissue continuity. This is secured by membrane-bound or diffusible signals that instruct the epithelial cells, in a coordinated fashion, to change shapes and adhesive properties and when, how and where to move. Here we show that during Dorsal Closure (DC) in Drosophila, the Jun kinase (JNK) signaling pathway modulates integrins expression and ensures tissue endurance. An excess of JNK activity, as an outcome of a failure in the negative feedback implemented by the dual-specificity phosphatase Puckered (Puc), promotes the loss of integrins [the ß-subunit Myospheroid (Mys)] and amnioserosa detachment. Likewise, integrins signal back to the pathway to regulate the duration and strength of JNK activity. Mys is necessary for the regulation of JNK activity levels and in its absence, puc expression is downregulated and JNK activity increases.
Collapse
Affiliation(s)
- Katerina Karkali
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| | - Jose Carlos Pastor-Pareja
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (IN-CSIC), Alicante, Spain
| | - Enrique Martin-Blanco
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| |
Collapse
|
3
|
The Tbx6 Transcription Factor Dorsocross Mediates Dpp Signaling to Regulate Drosophila Thorax Closure. Int J Mol Sci 2022; 23:ijms23094543. [PMID: 35562934 PMCID: PMC9104307 DOI: 10.3390/ijms23094543] [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: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/17/2022] [Indexed: 11/23/2022] Open
Abstract
Movement and fusion of separate cell populations are critical for several developmental processes, such as neural tube closure in vertebrates or embryonic dorsal closure and pupal thorax closure in Drosophila. Fusion failure results in an opening or groove on the body surface. Drosophila pupal thorax closure is an established model to investigate the mechanism of tissue closure. Here, we report the identification of T-box transcription factor genes Dorsocross (Doc) as Decapentaplegic (Dpp) targets in the leading edge cells of the notum in the late third instar larval and early pupal stages. Reduction of Doc in the notum region results in a thorax closure defect, similar to that in dpp loss-of-function flies. Nine genes are identified as potential downstream targets of Doc in regulating thorax closure by molecular and genetic screens. Our results reveal a novel function of Doc in Drosophila development. The candidate target genes provide new clues for unravelling the mechanism of collective cell movement.
Collapse
|
4
|
Karkali K, Martin-Blanco E. Dissection of the Regulatory Elements of the Complex Expression Pattern of Puckered, a Dual-Specificity JNK Phosphatase. Int J Mol Sci 2021; 22:ijms222212205. [PMID: 34830088 PMCID: PMC8623796 DOI: 10.3390/ijms222212205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
For developmental processes, we know most of the gene networks controlling specific cell responses. We still have to determine how these networks cooperate and how signals become integrated. The JNK pathway is one of the key elements modulating cellular responses during development. Yet, we still know little about how the core components of the pathway interact with additional regulators or how this network modulates cellular responses in the whole organism in homeostasis or during tissue morphogenesis. We have performed a promoter analysis, searching for potential regulatory sequences of puckered (puc) and identified different specific enhancers directing gene expression in different tissues and at different developmental times. Remarkably, some of these domains respond to the JNK activity, but not all. Altogether, these analyses show that puc expression regulation is very complex and that JNK activities participate in non-previously known processes during the development of Drosophila.
Collapse
|
5
|
Yoo B, Kim HY, Chen X, Shen W, Jang JS, Stein SN, Cormier O, Pereira L, Shih CRY, Krieger C, Reed B, Harden N, Wang SJH. 20-hydroxyecdysone (20E) signaling regulates amnioserosa morphogenesis during Drosophila dorsal closure: EcR modulates gene expression in a complex with the AP-1 subunit, Jun. Biol Open 2021; 10:271855. [PMID: 34296248 PMCID: PMC8411571 DOI: 10.1242/bio.058605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/06/2021] [Indexed: 11/25/2022] Open
Abstract
Steroid hormones influence diverse biological processes throughout the animal life cycle, including metabolism, stress resistance, reproduction, and lifespan. In insects, the steroid hormone, 20-hydroxyecdysone (20E), is the central hormone regulator of molting and metamorphosis, and plays roles in tissue morphogenesis. For example, amnioserosa contraction, which is a major driving force in Drosophila dorsal closure (DC), is defective in embryos mutant for 20E biosynthesis. Here, we show that 20E signaling modulates the transcription of several DC participants in the amnioserosa and other dorsal tissues during late embryonic development, including zipper, which encodes for non-muscle myosin. Canonical ecdysone signaling typically involves the binding of Ecdysone receptor (EcR) and Ultraspiracle heterodimers to ecdysone-response elements (EcREs) within the promoters of responsive genes to drive expression. During DC, however, we provide evidence that 20E signaling instead acts in parallel to the JNK cascade via a direct interaction between EcR and the AP-1 transcription factor subunit, Jun, which together binds to genomic regions containing AP-1 binding sites but no EcREs to control gene expression. Our work demonstrates a novel mode of action for 20E signaling in Drosophila that likely functions beyond DC, and may provide further insights into mammalian steroid hormone receptor interactions with AP-1. Summary: During Drosophila dorsal closure, 20E signaling acts non-canonically through an interaction between EcR and the AP-1 subunit, Jun, to control gene expression at regions containing AP-1 motifs but no EcREs.
Collapse
Affiliation(s)
- Byoungjoo Yoo
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Hae-Yoon Kim
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Xi Chen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Weiping Shen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Ji Sun Jang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Shaianne N Stein
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Olga Cormier
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Lionel Pereira
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Claire R Y Shih
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Charles Krieger
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Bruce Reed
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Nicholas Harden
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Simon J H Wang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada.,Department of Biomedical Physiology and Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
6
|
Morgani SM, Su J, Nichols J, Massagué J, Hadjantonakis AK. The transcription factor Rreb1 regulates epithelial architecture, invasiveness, and vasculogenesis in early mouse embryos. eLife 2021; 10:e64811. [PMID: 33929320 PMCID: PMC8131102 DOI: 10.7554/elife.64811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/16/2021] [Indexed: 12/23/2022] Open
Abstract
Ras-responsive element-binding protein 1 (Rreb1) is a zinc-finger transcription factor acting downstream of RAS signaling. Rreb1 has been implicated in cancer and Noonan-like RASopathies. However, little is known about its role in mammalian non-disease states. Here, we show that Rreb1 is essential for mouse embryonic development. Loss of Rreb1 led to a reduction in the expression of vasculogenic factors, cardiovascular defects, and embryonic lethality. During gastrulation, the absence of Rreb1 also resulted in the upregulation of cytoskeleton-associated genes, a change in the organization of F-ACTIN and adherens junctions within the pluripotent epiblast, and perturbed epithelial architecture. Moreover, Rreb1 mutant cells ectopically exited the epiblast epithelium through the underlying basement membrane, paralleling cell behaviors observed during metastasis. Thus, disentangling the function of Rreb1 in development should shed light on its role in cancer and other diseases involving loss of epithelial integrity.
Collapse
Affiliation(s)
- Sophie M Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Jeffrey Cheah Biomedical Centre Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Jie Su
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Jeffrey Cheah Biomedical Centre Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
7
|
Ng WA, Ma A, Chen M, Reed BH. A Method for Rapid Selection of Randomly Induced Mutations in a Gene of Interest Using CRISPR/Cas9 Mediated Activation of Gene Expression. G3 (BETHESDA, MD.) 2020; 10:1893-1901. [PMID: 32312838 PMCID: PMC7263667 DOI: 10.1534/g3.120.401299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/14/2020] [Indexed: 11/18/2022]
Abstract
We have developed a CRISPR/Cas9 based method for isolating randomly induced recessive lethal mutations in a gene of interest (GOI) by selection within the F1 progeny of a single genetic cross. Our method takes advantage of the ability to overexpress a GOI using CRISPR/Cas9 mediated activation of gene expression. In essence, the screening strategy is based upon the idea that if overexpression of a wild type allele can generate a phenotype, then overexpression of a newly induced loss-of-function allele will lack this phenotype. As a proof-of-principle, we used this method to select EMS induced mutations of the Drosophila gene hindsight (hnt). From approximately 45,000 F1 progeny we recovered 8 new EMS induced loss-of-function hnt alleles that we characterized as an allelic series of hypomorphic mutations. This new method can, in theory, be used to recover randomly induced point mutants in a GOI and can be applied to any circumstance where CRISPR/Cas9 mediated activation of gene expression is associated with lethality or a visible phenotype.
Collapse
Affiliation(s)
- William A Ng
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Andrew Ma
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Molly Chen
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Bruce H Reed
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| |
Collapse
|
8
|
A Functional Analysis of the Drosophila Gene hindsight: Evidence for Positive Regulation of EGFR Signaling. G3-GENES GENOMES GENETICS 2020; 10:117-127. [PMID: 31649045 PMCID: PMC6945037 DOI: 10.1534/g3.119.400829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have investigated the relationship between the function of the gene hindsight (hnt), which is the Drosophila homolog of Ras Responsive Element Binding protein-1 (RREB-1), and the EGFR signaling pathway. We report that hnt mutant embryos are defective in EGFR signaling dependent processes, namely chordotonal organ recruitment and oenocyte specification. We also show the temperature sensitive hypomorphic allele hntpebbled is enhanced by the hypomorphic MAPK allele rolled (rl1 ). We find that hnt overexpression results in ectopic DPax2 expression within the embryonic peripheral nervous system, and we show that this effect is EGFR-dependent. Finally, we show that the canonical U-shaped embryonic lethal phenotype of hnt, which is associated with premature degeneration of the extraembyonic amnioserosa and a failure in germ band retraction, is rescued by expression of several components of the EGFR signaling pathway (sSpi, Ras85D V12 , pntP1 ) as well as the caspase inhibitor p35 Based on this collection of corroborating evidence, we suggest that an overarching function of hnt involves the positive regulation of EGFR signaling.
Collapse
|
9
|
Alfonso-Gonzalez C, Riesgo-Escovar JR. Fos metamorphoses: Lessons from mutants in model organisms. Mech Dev 2018; 154:73-81. [PMID: 29753813 DOI: 10.1016/j.mod.2018.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
The Fos oncogene gene family is evolutionarily conserved throughout Eukarya. Fos proteins characteristically have a leucine zipper and a basic region with a helix-turn-helix motif that binds DNA. In vertebrates, there are several Fos homologs. They can homo- or hetero-dimerize via the leucine zipper domain. Fos homologs coupled with other transcription factors, like Jun oncoproteins, constitute the Activator Protein 1 (AP-1) complex. From its original inception as an oncogene, the subsequent finding that they act as transcription factors binding DNA sequences known as TRE, to the realization that they are activated in many different scenarios, and to loss-of-function analysis, the Fos proteins have traversed a multifarious path in development and physiology. They are instrumental in 'immediate early genes' responses, and activated by a seemingly myriad assemblage of different stimuli. Yet, the majority of these studies were basically gain-of-function studies, since it was thought that Fos genes would be cell lethal. Loss-of-function mutations in vertebrates were recovered later, and were not cell lethal. In fact, c-fos null mutations are viable with developmental defects (osteopetrosis and myeloid lineage abnormalities). It was then hypothesized that vertebrate genomes exhibit partial redundancy, explaining the 'mild' phenotypes, and complicating assessment of complete loss-of-function phenotypes. Due to its promiscuous activation, fos genes (especially c-fos) are now commonly used as markers for cellular responses to stimuli. fos homologs high sequence conservation (including Drosophila) is advantageous as it allows critical assessment of fos genes functions in this genetic model. Drosophila melanogaster contains only one fos homolog, the gene kayak. kayak mutations are lethal, and allow study of all the processes where fos is required. The kayak locus encodes several different isoforms, and is a pleiotropic gene variously required for development involving cell shape changes. In general, fos genes seem to primarily activate programs involved in cellular architectural rearrangements and cell shape changes.
Collapse
Affiliation(s)
- Carlos Alfonso-Gonzalez
- Developmental Neurobiology and Neurophysiology Department, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro c.p.76230, Mexico; Maestría en Bioquímica y Biología Molecular, Facultad de Química, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Juan Rafael Riesgo-Escovar
- Developmental Neurobiology and Neurophysiology Department, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro c.p.76230, Mexico.
| |
Collapse
|
10
|
Kiehart DP, Crawford JM, Aristotelous A, Venakides S, Edwards GS. Cell Sheet Morphogenesis: Dorsal Closure in Drosophila melanogaster as a Model System. Annu Rev Cell Dev Biol 2018; 33:169-202. [PMID: 28992442 DOI: 10.1146/annurev-cellbio-111315-125357] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Dorsal closure is a key process during Drosophila morphogenesis that models cell sheet movements in chordates, including neural tube closure, palate formation, and wound healing. Closure occurs midway through embryogenesis and entails circumferential elongation of lateral epidermal cell sheets that close a dorsal hole filled with amnioserosa cells. Signaling pathways regulate the function of cellular structures and processes, including Actomyosin and microtubule cytoskeletons, cell-cell/cell-matrix adhesion complexes, and endocytosis/vesicle trafficking. These orchestrate complex shape changes and movements that entail interactions between five distinct cell types. Genetic and laser perturbation studies establish that closure is robust, resilient, and the consequence of redundancy that contributes to four distinct biophysical processes: contraction of the amnioserosa, contraction of supracellular Actomyosin cables, elongation (stretching?) of the lateral epidermis, and zipping together of two converging cell sheets. What triggers closure and what the emergent properties are that give rise to its extraordinary resilience and fidelity remain key, extant questions.
Collapse
Affiliation(s)
- Daniel P Kiehart
- Department of Biology, Duke University, Durham, North Carolina 27708;
| | - Janice M Crawford
- Department of Biology, Duke University, Durham, North Carolina 27708;
| | - Andreas Aristotelous
- Department of Mathematics, West Chester University, West Chester, Pennsylvania 19383
| | | | - Glenn S Edwards
- Physics Department, Duke University, Durham, North Carolina 27708
| |
Collapse
|
11
|
Farley JE, Burdett TC, Barria R, Neukomm LJ, Kenna KP, Landers JE, Freeman MR. Transcription factor Pebbled/RREB1 regulates injury-induced axon degeneration. Proc Natl Acad Sci U S A 2018; 115:1358-1363. [PMID: 29295933 PMCID: PMC5819420 DOI: 10.1073/pnas.1715837115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genetic studies of Wallerian degeneration have led to the identification of signaling molecules (e.g., dSarm/Sarm1, Axundead, and Highwire) that function locally in axons to drive degeneration. Here we identify a role for the Drosophila C2H2 zinc finger transcription factor Pebbled [Peb, Ras-responsive element binding protein 1 (RREB1) in mammals] in axon death. Loss of Peb in Drosophila glutamatergic sensory neurons results in either complete preservation of severed axons, or an axon death phenotype where axons fragment into large, continuous segments, rather than completely disintegrate. Peb is expressed in developing and mature sensory neurons, suggesting it is required to establish or maintain their competence to undergo axon death. peb mutant phenotypes can be rescued by human RREB1, and they exhibit dominant genetic interactions with dsarm mutants, linking peb/RREB1 to the axon death signaling cascade. Surprisingly, Peb is only able to fully block axon death signaling in glutamatergic, but not cholinergic sensory neurons, arguing for genetic diversity in axon death signaling programs in different neuronal subtypes. Our findings identify a transcription factor that regulates axon death signaling, and peb mutant phenotypes of partial fragmentation reveal a genetically accessible step in axon death signaling.
Collapse
Affiliation(s)
- Jonathan E Farley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Thomas C Burdett
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Romina Barria
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Lukas J Neukomm
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Kevin P Kenna
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655;
| |
Collapse
|
12
|
Deady LD, Li W, Sun J. The zinc-finger transcription factor Hindsight regulates ovulation competency of Drosophila follicles. eLife 2017; 6:29887. [PMID: 29256860 PMCID: PMC5768419 DOI: 10.7554/elife.29887] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/18/2017] [Indexed: 12/15/2022] Open
Abstract
Follicle rupture, the final step in ovulation, utilizes conserved molecular mechanisms including matrix metalloproteinases (Mmps), steroid signaling, and adrenergic signaling. It is still unknown how follicles become competent for follicle rupture/ovulation. Here, we identify a zinc-finger transcription factor Hindsight (Hnt) as the first transcription factor regulating follicle’s competency for ovulation in Drosophila. Hnt is not expressed in immature stage-13 follicle cells but is upregulated in mature stage-14 follicle cells, which is essential for follicle rupture/ovulation. Hnt upregulates Mmp2 expression in posterior follicle cells (essential for the breakdown of the follicle wall) and Oamb expression in all follicle cells (the receptor for receiving adrenergic signaling and inducing Mmp2 activation). Hnt’s role in regulating Mmp2 and Oamb can be replaced by its human homolog Ras-responsive element-binding protein 1 (RREB-1). Our data suggest that Hnt/RREB-1 plays conserved role in regulating follicle maturation and competency for ovulation. The release of an egg from the ovary of a female animal is a process known as ovulation. Animals as different as humans and fruit flies ovulate in largely similar ways. Yet the systems involved in controlling ovulation are still not well understood. An egg cell develops within a collection of cells that help the egg to form properly. Together, this unit is called a follicle. During ovulation, connections between the egg and the rest of the follicle break down and the egg is eventually ejected. Ovulation happens in response to a hormone signal from the brain. In humans, this hormone is called luteinizing hormone, whereas in flies it is called octopamine. Specialized protein molecules on the surface of the follicle cells receive these hormone signals, but can only cause ovulation in mature follicles. It was not clear what allows only mature follicles to ovulate. Deady et al. have now used the fruit fly Drosophila melanogaster to examine ovulation to identify how the process is controlled. The results showed that a protein called Hindsight primes follicle cells for ovulation. When a follicle reaches its final stage (called stage 14 in flies), the gene for Hindsight becomes active and produces the protein. This protein then activates other genes. One of the activated genes makes a protein that receives the hormone signal, while another makes a protein that breaks down follicle cells and allows the egg to be released. The findings of Deady et al. reveal that Hindsight is needed for ovulation in flies. Further experiments then showed that the gene for equivalent human protein can be transplanted into flies and can still prime follicles for ovulation. This indicates that the genes in humans and flies may perform the same tasks. Studying ovulation is an important part of understanding female fertility and could help scientists to understand more about human reproduction. These results may also lead to new contraceptives and improved approaches for treating infertility.
Collapse
Affiliation(s)
- Lylah D Deady
- Department of Physiology and Neurobiology, University of Connecticut, Connecticut, United States
| | - Wei Li
- Department of Physiology and Neurobiology, University of Connecticut, Connecticut, United States
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Connecticut, United States.,Institute for Systems Genomics, University of Connecticut, Connecticut, United States
| |
Collapse
|
13
|
Studying Nonproliferative Roles for Egfr Signaling in Tissue Morphogenesis Using Dorsal Closure of the Drosophila Embryo. Methods Mol Biol 2017. [PMID: 28791646 DOI: 10.1007/978-1-4939-7219-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
For several decades, genetic analysis in Drosophila has made important contributions to the understanding of signaling by Egfr. Egfr has been well characterized with regard to its oncogenic potential but is also being studied for its roles in organismal development. We have recently developed dorsal closure of the Drosophila embryo as a system for characterizing Egfr regulation of events that do not involve proliferation, as no cell divisions occur during this process. Dorsal closure is essentially a developmental wound healing event with parallels to vertebrate developmental epithelial fusions such as neural tube closure and palate fusion. We describe here a set of materials and protocols for studying Egfr signaling during dorsal closure, including assessing effects of altering Egfr signaling on other pathways, gene expression and, using live imaging, morphogenesis and programmed cell death. Although this "tool kit" is designed for looking at Egfr, it can be readily adapted to look at the participation of any signaling molecule in dorsal closure.
Collapse
|
14
|
Treffkorn S, Mayer G. Conserved versus derived patterns of controlled cell death during the embryonic development of two species of Onychophora (velvet worms). Dev Dyn 2017; 246:403-416. [PMID: 28198063 DOI: 10.1002/dvdy.24492] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Apoptosis is involved in various developmental processes, including cell migration and tissue and organ formation. Some of these processes are conserved across metazoans, while others are specific to particular taxa. Although the patterns of apoptosis have been investigated in arthropods, no corresponding data are available from one of their closest relatives, the Onychophora (velvet worms). RESULTS We analyzed the patterns of apoptosis in embryos of two onychophoran species: the lecithotrophic/matrotrophic viviparous peripatopsid Euperipatoides rowelli, and the placentotrophic viviparous peripatid Principapillatus hitoyensis. Our data show that apoptosis occurs early in development and might be responsible for the degeneration of extra-embryonic tissues. Moreover, apoptosis might be involved in the morphogenesis of the ventral and preventral organs in both species and occurs additionally in the placental stalk of P. hitoyensis. CONCLUSIONS Despite the different developmental modes in these onychophoran species, our data suggest that patterns of apoptosis are conserved among onychophorans. While apoptosis in the dorsal extra-embryonic tissue might contribute to dorsal closure-a process also known from arthropods-the involvement of apoptosis in ventral closure might be unique to onychophorans. Apoptosis in the placental stalk of P. hitoyensis is most likely a derived feature of the placentotrophic onychophorans. Developmental Dynamics 246:403-416, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sandra Treffkorn
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| | - Georg Mayer
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| |
Collapse
|
15
|
Hayes P, Solon J. Drosophila dorsal closure: An orchestra of forces to zip shut the embryo. Mech Dev 2017; 144:2-10. [PMID: 28077304 DOI: 10.1016/j.mod.2016.12.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/17/2016] [Accepted: 12/23/2016] [Indexed: 10/20/2022]
Abstract
Dorsal closure, a late-embryogenesis process, consists in the sealing of an epidermal gap on the dorsal side of the Drosophila embryo. Because of its similarities with wound healing and neural tube closure in humans, it has been extensively studied in the last twenty years. The process requires the coordination of several force generating mechanisms, that together will zip shut the epidermis. Recent works have provided a precise description of the cellular behavior at the origin of these forces and proposed quantitative models of the process. In this review, we will describe the different forces acting in dorsal closure. We will present our current knowledge on the mechanisms generating and regulating these forces and report on the different quantitative mathematical models proposed so far.
Collapse
Affiliation(s)
- Peran Hayes
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain
| | - Jérôme Solon
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain.
| |
Collapse
|
16
|
Baechler BL, McKnight C, Pruchnicki PC, Biro NA, Reed BH. Hindsight/RREB-1 functions in both the specification and differentiation of stem cells in the adult midgut of Drosophila. Biol Open 2015; 5:1-10. [PMID: 26658272 PMCID: PMC4728307 DOI: 10.1242/bio.015636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The adult Drosophila midgut is established during the larval/pupal transition from undifferentiated cells known as adult midgut precursors (AMPs). Four fundamental cell types are found in the adult midgut epithelium: undifferentiated intestinal stem cells (ISCs) and their committed daughter cells, enteroblasts (EBs), plus enterocytes (ECs) and enteroendocrine cells (EEs). Using the Drosophila posterior midgut as a model, we have studied the function of the transcription factor Hindsight (Hnt)/RREB-1 and its relationship to the Notch and Egfr signaling pathways. We show that hnt is required for EC differentiation in the context of ISC-to-EC differentiation, but not in the context of AMP-to-EC differentiation. In addition, we show that hnt is required for the establishment of viable or functional ISCs. Overall, our studies introduce hnt as a key factor in the regulation of both the developing and the mature adult midgut. We suggest that the nature of these contextual differences can be explained through the interaction of hnt with multiple signaling pathways.
Collapse
Affiliation(s)
- Brittany L Baechler
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Cameron McKnight
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Porsha C Pruchnicki
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Nicole A Biro
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Bruce H Reed
- Department of Biology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
17
|
Flores-Benitez D, Knust E. Crumbs is an essential regulator of cytoskeletal dynamics and cell-cell adhesion during dorsal closure in Drosophila. eLife 2015; 4. [PMID: 26544546 PMCID: PMC4718732 DOI: 10.7554/elife.07398] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved Crumbs protein is required for epithelial polarity and morphogenesis. Here we identify a novel role of Crumbs as a negative regulator of actomyosin dynamics during dorsal closure in the Drosophila embryo. Embryos carrying a mutation in the FERM (protein 4.1/ezrin/radixin/moesin) domain-binding motif of Crumbs die due to an overactive actomyosin network associated with disrupted adherens junctions. This phenotype is restricted to the amnioserosa and does not affect other embryonic epithelia. This function of Crumbs requires DMoesin, the Rho1-GTPase, class-I p21-activated kinases and the Arp2/3 complex. Data presented here point to a critical role of Crumbs in regulating actomyosin dynamics, cell junctions and morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.001 A layer of epithelial cells covers the body surface of animals. Epithelial cells have a property known as polarity; this means that they have two different poles, one of which is in contact with the environment. Midway through embryonic development, the Drosophila embryo is covered by two kinds of epithelial sheets; the epidermis on the front, the belly and the sides of the embryo, and the amnioserosa on the back. In the second half of embryonic development, the amnioserosa is brought into the embryo in a process called dorsal closure, while the epidermis expands around the back of the embryo to encompass it. One of the major activities driving dorsal closure is the contraction of amnioserosa cells. This contraction depends on the highly dynamic activity of the protein network that helps give cells their shape, known as the actomyosin cytoskeleton. One major question in the field is how changes in the actomyosin cytoskeleton are controlled as tissues take shape (a process known as “morphogenesis”) and how the integrity of epithelial tissues is maintained during these processes. A key regulator of epidermal and amnioserosa polarity is an evolutionarily conserved protein called Crumbs. The epithelial tissues of mutant embryos that do not produce Crumbs lose polarity and integrity, and the embryos fail to develop properly. Flores-Benitez and Knust have now studied the role of Crumbs in the morphogenesis of the amnioserosa during dorsal closure. This revealed that fly embryos that produce a mutant Crumbs protein that cannot interact with a protein called Moesin (which links the cell membrane and the actomyosin cytoskeleton) are unable to complete dorsal closure. Detailed analyses showed that this failure of dorsal closure is due to the over-activity of the actomyosin cytoskeleton in the amnioserosa. This results in increased and uncoordinated contractions of the cells, and is accompanied by defects in cell-cell adhesion that ultimately cause the amnioserosa to lose integrity. Flores-Benitez and Knust’s genetic analyses further showed that several different signalling systems participate in this process. Flores-Benitez and Knust’s results reveal an unexpected role of Crumbs in coordinating polarity, actomyosin activity and cell-cell adhesion. Further work is now needed to understand the molecular mechanisms and interactions that enable Crumbs to coordinate these processes; in particular, to unravel how Crumbs influences the periodic contractions that drive changes in cell shape. It will also be important to investigate whether Crumbs is involved in similar mechanisms that operate in other developmental events in which actomyosin oscillations have been linked to tissue morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.002
Collapse
Affiliation(s)
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
18
|
acal is a long non-coding RNA in JNK signaling in epithelial shape changes during drosophila dorsal closure. PLoS Genet 2015; 11:e1004927. [PMID: 25710168 PMCID: PMC4339196 DOI: 10.1371/journal.pgen.1004927] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 12/01/2014] [Indexed: 11/19/2022] Open
Abstract
Dorsal closure is an epithelial remodeling process taking place during Drosophila embryogenesis. JNK signaling coordinates dorsal closure. We identify and characterize acal as a novel negative dorsal closure regulator. acal represents a new level of JNK regulation. The acal locus codes for a conserved, long, non-coding, nuclear RNA. Long non-coding RNAs are an abundant and diverse class of gene regulators. Mutations in acal are lethal. acal mRNA expression is dynamic and is processed into a collection of 50 to 120 bp fragments. We show that acal lies downstream of raw, a pioneer protein, helping explain part of raw functions, and interacts genetically with Polycomb. acal functions in trans regulating mRNA expression of two genes involved in JNK signaling and dorsal closure: Connector of kinase to AP1 (Cka) and anterior open (aop). Cka is a conserved scaffold protein that brings together JNK and Jun, and aop is a transcription factor. Misregulation of Cka and aop can account for dorsal closure phenotypes in acal mutants. Changes in cell shape affect many critical cellular and bodily processes, like wound healing and developmental events, and when gone awry, metastatic processes in cancer. Evolutionarily conserved signaling pathways govern regulation of these cellular changes. The Jun-N-terminal kinase pathway regulates cell stretching during wound healing and normal development. An extensively studied developmental process is embryonic dorsal closure in fruit flies, a well-established model for the regulation and manner of this cell shape changes. Here we describe and characterize a processed, long non-coding RNA locus, acal, that adds a new layer of complexity to the Jun-N-terminal kinase signaling, acting as a negative regulator of the pathway. acal modulates the expression of two key genes in the pathway: the scaffold protein Cka, and the transcription factor Aop. Together, they enable the proper level of Jun-N-terminal kinase pathway activation to occur to allow cell stretching and closure.
Collapse
|
19
|
Oliva C, Molina-Fernandez C, Maureira M, Candia N, López E, Hassan B, Aerts S, Cánovas J, Olguín P, Sierralta J. Hindsight regulates photoreceptor axon targeting through transcriptional control of jitterbug/Filamin and multiple genes involved in axon guidance in Drosophila. Dev Neurobiol 2015; 75:1018-32. [PMID: 25652545 DOI: 10.1002/dneu.22271] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 01/20/2023]
Abstract
During axon targeting, a stereotyped pattern of connectivity is achieved by the integration of intrinsic genetic programs and the response to extrinsic long and short-range directional cues. How this coordination occurs is the subject of intense study. Transcription factors play a central role due to their ability to regulate the expression of multiple genes required to sense and respond to these cues during development. Here we show that the transcription factor HNT regulates layer-specific photoreceptor axon targeting in Drosophila through transcriptional control of jbug/Filamin and multiple genes involved in axon guidance and cytoskeleton organization.Using a microarray analysis we identified 235 genes whose expression levels were changed by HNT overexpression in the eye primordia. We analyzed nine candidate genes involved in cytoskeleton regulation and axon guidance, six of which displayed significantly altered gene expression levels in hnt mutant retinas. Functional analysis confirmed the role of OTK/PTK7 in photoreceptor axon targeting and uncovered Tiggrin, an integrin ligand, and Jbug/Filamin, a conserved actin- binding protein, as new factors that participate of photoreceptor axon targeting. Moreover, we provided in silico and molecular evidence that supports jbug/Filamin as a direct transcriptional target of HNT and that HNT acts partially through Jbug/Filamin in vivo to regulate axon guidance. Our work broadens the understanding of how HNT regulates the coordinated expression of a group of genes to achieve the correct connectivity pattern in the Drosophila visual system. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 75: 1018-1032, 2015.
Collapse
Affiliation(s)
- Carlos Oliva
- Laboratorio de Neurobiología Celular y Molecular, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile.,Biomedical Neurosciences Institute, ICM, Facultad de Medicina, Universidad de Chile
| | - Claudia Molina-Fernandez
- Laboratorio de Genética del Desarrollo de Drosophila, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile
| | - Miguel Maureira
- Laboratorio de Genética del Desarrollo de Drosophila, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile
| | - Noemi Candia
- Laboratorio de Genética del Desarrollo de Drosophila, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile
| | - Estefanía López
- Laboratorio de Neurobiología Celular y Molecular, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile.,Biomedical Neurosciences Institute, ICM, Facultad de Medicina, Universidad de Chile
| | - Bassem Hassan
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, VIB, K.U. Leuven, Leuven, Belgium
| | - Stein Aerts
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, VIB, K.U. Leuven, Leuven, Belgium
| | - José Cánovas
- Laboratorio de Neurobiología Celular y Molecular, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile.,Biomedical Neurosciences Institute, ICM, Facultad de Medicina, Universidad de Chile
| | - Patricio Olguín
- Laboratorio de Genética del Desarrollo de Drosophila, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile
| | - Jimena Sierralta
- Laboratorio de Neurobiología Celular y Molecular, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile.,Biomedical Neurosciences Institute, ICM, Facultad de Medicina, Universidad de Chile
| |
Collapse
|
20
|
Nowotarski SH, McKeon N, Moser RJ, Peifer M. The actin regulators Enabled and Diaphanous direct distinct protrusive behaviors in different tissues during Drosophila development. Mol Biol Cell 2014; 25:3147-65. [PMID: 25143400 PMCID: PMC4196866 DOI: 10.1091/mbc.e14-05-0951] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Actin-based protrusions are important for signaling and migration during development and homeostasis. Gain- and loss-of-function and quantitative approaches are used to define differential roles for the actin elongation factors Diaphanous and Enabled in regulating distinct protrusive behaviors in different tissues during Drosophila morphogenesis. Actin-based protrusions are important for signaling and migration during development and homeostasis. Defining how different tissues in vivo craft diverse protrusive behaviors using the same genomic toolkit of actin regulators is a current challenge. The actin elongation factors Diaphanous and Enabled both promote barbed-end actin polymerization and can stimulate filopodia in cultured cells. However, redundancy in mammals and Diaphanous’ role in cytokinesis limited analysis of whether and how they regulate protrusions during development. We used two tissues driving Drosophila dorsal closure—migratory leading-edge (LE) and nonmigratory amnioserosal (AS) cells—as models to define how cells shape distinct protrusions during morphogenesis. We found that nonmigratory AS cells produce filopodia that are morphologically and dynamically distinct from those of LE cells. We hypothesized that differing Enabled and/or Diaphanous activity drives these differences. Combining gain- and loss-of-function with quantitative approaches revealed that Diaphanous and Enabled each regulate filopodial behavior in vivo and defined a quantitative “fingerprint”—the protrusive profile—which our data suggest is characteristic of each actin regulator. Our data suggest that LE protrusiveness is primarily Enabled driven, whereas Diaphanous plays the primary role in the AS, and reveal each has roles in dorsal closure, but its robustness ensures timely completion in their absence.
Collapse
Affiliation(s)
- Stephanie H Nowotarski
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natalie McKeon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Rachel J Moser
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Mark Peifer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
21
|
Ming L, Wilk R, Reed BH, Lipshitz HD. Drosophila Hindsight and mammalian RREB-1 are evolutionarily conserved DNA-binding transcriptional attenuators. Differentiation 2014; 86:159-70. [PMID: 24418439 DOI: 10.1016/j.diff.2013.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/25/2013] [Accepted: 12/05/2013] [Indexed: 12/22/2022]
Abstract
The Drosophila Hindsight (hnt) gene encodes a C2H2-type Zinc-finger protein, HNT, that plays multiple developmental roles including control of embryonic germ band retraction and regulation of retinal cell fate and morphogenesis. While the developmental functions of the human HNT homolog, RREB-1, are unknown, it has been shown to function as a transcriptional modulator of several tumor suppressor genes. Here we investigate HNT's functional motifs, target genes and its regulatory abilities. We show that the C-terminal region of HNT, containing the last five of its 14 Zinc fingers, binds in vitro to DNA elements very similar to those identified for RREB-1. We map HNT's in vivo binding sites on salivary gland polytene chromosomes and define, at high resolution, where HNT is bound to two target genes, hnt itself and nervy (nvy). Data from both loss-of-function and over-expression experiments show that HNT attenuates the transcription of these two targets in a tissue-specific manner. RREB-1, when expressed in Drosophila, binds to the same polytene chromosome sites as HNT, attenuates expression of the hnt and nvy genes, and rescues the germ band retraction phenotype. HNT's ninth Zinc finger has degenerated or been lost in the vertebrate lineage. We show that a HNT protein mutant for this finger can also attenuate target gene expression and rescue germ band retraction. Thus HNT and RREB-1 are functional homologs at the level of DNA binding, transcriptional regulation and developmental control.
Collapse
Affiliation(s)
- Liang Ming
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8.
| | - Ronit Wilk
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8.
| | - Bruce H Reed
- Department of Biology, University of Waterloo, 200 University Avenue W, Waterloo, Ontario, Canada N2L 3G1.
| | - Howard D Lipshitz
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8.
| |
Collapse
|
22
|
Shen W, Chen X, Cormier O, Cheng DCP, Reed B, Harden N. Modulation of morphogenesis by Egfr during dorsal closure in Drosophila. PLoS One 2013; 8:e60180. [PMID: 23579691 PMCID: PMC3620322 DOI: 10.1371/journal.pone.0060180] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/23/2013] [Indexed: 01/12/2023] Open
Abstract
During Drosophila embryogenesis the process of dorsal closure (DC) results in continuity of the embryonic epidermis, and DC is well recognized as a model system for the analysis of epithelial morphogenesis as well as wound healing. During DC the flanking lateral epidermal sheets stretch, align, and fuse along the dorsal midline, thereby sealing a hole in the epidermis occupied by an extra-embryonic tissue known as the amnioserosa (AS). Successful DC requires the regulation of cell shape change via actomyosin contractility in both the epidermis and the AS, and this involves bidirectional communication between these two tissues. We previously demonstrated that transcriptional regulation of myosin from the zipper (zip) locus in both the epidermis and the AS involves the expression of Ack family tyrosine kinases in the AS in conjunction with Dpp secreted from the epidermis. A major function of Ack in other species, however, involves the negative regulation of Egfr. We have, therefore, asked what role Egfr might play in the regulation of DC. Our studies demonstrate that Egfr is required to negatively regulate epidermal expression of dpp during DC. Interestingly, we also find that Egfr signaling in the AS is required to repress zip expression in both the AS and the epidermis, and this may be generally restrictive to the progression of morphogenesis in these tissues. Consistent with this theme of restricting morphogenesis, it has previously been shown that programmed cell death of the AS is essential for proper DC, and we show that Egfr signaling also functions to inhibit or delay AS programmed cell death. Finally, we present evidence that Ack regulates zip expression by promoting the endocytosis of Egfr in the AS. We propose that the general role of Egfr signaling during DC is that of a braking mechanism on the overall progression of DC.
Collapse
Affiliation(s)
- Weiping Shen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Xi Chen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Olga Cormier
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - David Chung-Pei Cheng
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Bruce Reed
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Nicholas Harden
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
23
|
Langen M, Koch M, Yan J, De Geest N, Erfurth ML, Pfeiffer BD, Schmucker D, Moreau Y, Hassan BA. Mutual inhibition among postmitotic neurons regulates robustness of brain wiring in Drosophila. eLife 2013; 2:e00337. [PMID: 23471010 PMCID: PMC3589824 DOI: 10.7554/elife.00337] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 01/22/2013] [Indexed: 11/13/2022] Open
Abstract
Brain connectivity maps display a delicate balance between individual variation and stereotypy, suggesting the existence of dedicated mechanisms that simultaneously permit and limit individual variation. We show that during the development of the Drosophila central nervous system, mutual inhibition among groups of neighboring postmitotic neurons during development regulates the robustness of axon target choice in a nondeterministic neuronal circuit. Specifically, neighboring postmitotic neurons communicate through Notch signaling during axonal targeting, to ensure balanced alternative axon target choices without a corresponding change in cell fate. Loss of Notch in postmitotic neurons modulates an axon's target choice. However, because neighboring axons respond by choosing the complementary target, the stereotyped connectivity pattern is preserved. In contrast, loss of Notch in clones of neighboring postmitotic neurons results in erroneous coinnervation by multiple axons. Our observations establish mutual inhibition of axonal target choice as a robustness mechanism for brain wiring and unveil a novel cell fate independent function for canonical Notch signaling. DOI:http://dx.doi.org/10.7554/eLife.00337.001.
Collapse
Affiliation(s)
- Marion Langen
- Center for the Biology of Disease , VIB , Leuven , Belgium ; Center for Human Genetics , University of Leuven School of Medicine , Leuven , Belgium ; Doctoral Program in Molecular and Cognitive Neuroscience, Doctoral School of Biomedical Sciences , University of Leuven , Leuven , Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ríos-Barrera LD, Riesgo-Escovar JR. Regulating cell morphogenesis: The drosophila jun N-terminal kinase pathway. Genesis 2012; 51:147-62. [DOI: 10.1002/dvg.22354] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 10/14/2012] [Accepted: 10/19/2012] [Indexed: 12/15/2022]
Affiliation(s)
- Luis Daniel Ríos-Barrera
- Developmental Neurobioloy and Neurophysiology Department; Instituto de Neurobiología; Universidad Nacional Autónoma de México; Boulevard Juriquilla #3001; Querétaro, Querétaro; México; c.p. 76230
| | - Juan Rafael Riesgo-Escovar
- Developmental Neurobioloy and Neurophysiology Department; Instituto de Neurobiología; Universidad Nacional Autónoma de México; Boulevard Juriquilla #3001; Querétaro, Querétaro; México; c.p. 76230
| |
Collapse
|
25
|
Lada K, Gorfinkiel N, Martinez Arias A. Interactions between the amnioserosa and the epidermis revealed by the function of the u-shaped gene. Biol Open 2012; 1:353-61. [PMID: 23213425 PMCID: PMC3509461 DOI: 10.1242/bio.2012497] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dorsal closure (DC) is an essential step during Drosophila development whereby a hole is sealed in the dorsal epidermis and serves as a model for cell sheet morphogenesis and wound healing. It involves the orchestrated interplay of transcriptional networks and dynamic regulation of cell machinery to bring about shape changes, mechanical forces, and emergent properties. Here we provide insight into the regulation of dorsal closure by describing novel autonomous and non-autonomous roles for U-shaped (Ush) in the amnioserosa, the epidermis, and in mediation of communication between the tissues. We identified Ush by gene expression microarray analysis of Dpp signaling targets and show that Ush mediates some DC functions of Dpp. By selectively restoring Ush function in either the AS or the epidermis in ush mutants, we show that the AS makes a greater (Ush-dependent) contribution to closure than the epidermis. A signal from the AS induces epidermal cell elongation and JNK activation in the DME, while cable formation requires Ush on both sides of the leading edge, i.e. in both the AS and epidermis. Our study demonstrates that the amnioserosa and epidermis communicate at several steps during the process: sometimes the epidermis instructs the amnioserosa, other times the AS instructs the epidermis, and still other times they appear to collaborate.
Collapse
Affiliation(s)
- Karolina Lada
- Department of Genetics, University of Cambridge , CB2 3EH, Cambridge , UK
| | | | | |
Collapse
|
26
|
Belacortu Y, Paricio N. Drosophila as a model of wound healing and tissue regeneration in vertebrates. Dev Dyn 2011; 240:2379-404. [PMID: 21953647 DOI: 10.1002/dvdy.22753] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2011] [Indexed: 11/11/2022] Open
Abstract
Understanding the molecular basis of wound healing and regeneration in vertebrates is one of the main challenges in biology and medicine. This understanding will lead to medical advances allowing accelerated tissue repair after wounding, rebuilding new tissues/organs and restoring homeostasis. Drosophila has emerged as a valuable model for studying these processes because the genetic networks and cytoskeletal machinery involved in epithelial movements occurring during embryonic dorsal closure, larval imaginal disc fusion/regeneration, and epithelial repair are similar to those acting during wound healing and regeneration in vertebrates. Recent studies have also focused on the use of Drosophila adult stem cells to maintain tissue homeostasis. Here, we review how Drosophila has contributed to our understanding of these processes, primarily through live-imaging and genetic tools that are impractical in mammals. Furthermore, we highlight future research areas where this insect may provide novel insights and potential therapeutic strategies for wound healing and regeneration.
Collapse
Affiliation(s)
- Yaiza Belacortu
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, Burjasot, Spain
| | | |
Collapse
|
27
|
Galy A, Schenck A, Sahin HB, Qurashi A, Sahel JA, Diebold C, Giangrande A. CYFIP dependent actin remodeling controls specific aspects of Drosophila eye morphogenesis. Dev Biol 2011; 359:37-46. [PMID: 21884694 DOI: 10.1016/j.ydbio.2011.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 08/11/2011] [Accepted: 08/15/2011] [Indexed: 12/20/2022]
Abstract
Cell rearrangements shape organs and organisms using molecular pathways and cellular processes that are still poorly understood. Here we investigate the role of the Actin cytoskeleton in the formation of the Drosophila compound eye, which requires extensive remodeling and coordination between different cell types. We show that CYFIP/Sra-1, a member of the WAVE/SCAR complex and regulator of Actin remodeling, controls specific aspects of eye architecture: rhabdomere extension, rhabdomere terminal web organization, adherens junctions, retina depth and basement membrane integrity. We demonstrate that some phenotypes manifest independently, due to defects in different cell types. Mutations in WAVE/SCAR and in ARP2/3 complex subunits but not in WASP, another major regulator of Actin nucleation, phenocopy CYFIP defects. Thus, the CYFIP-SCAR-ARP2/3 pathway orchestrates specific tissue remodeling processes.
Collapse
Affiliation(s)
- Anne Galy
- Fovea-Pharmaceuticals, Institut de la vision, 17 rue Moreau 75012 Paris, France
| | - Annette Schenck
- Department of Human Genetics, Nijmegen Centre for Molecular Life Sciences, Donders Institute for Brain, Cognition and Behaviour & Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - H Bahar Sahin
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, Turkey
| | - Abrar Qurashi
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - José-Alain Sahel
- INSERM, U968, Université Pierre et Marie Curie-Paris 6, UM80, Institut de la Vision, CNRS, UMR-7210, Fondation Ophtalmologique Adolphe de Rothschild, Paris, France; Institute of Ophthalmology, University College of London, London, United Kingdom
| | - Céline Diebold
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, BP 10142, 67404 Illkirch, CU de Strasbourg, France
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, BP 10142, 67404 Illkirch, CU de Strasbourg, France.
| |
Collapse
|
28
|
Wang X, Ward RE. Sec61alpha is required for dorsal closure during Drosophila embryogenesis through its regulation of Dpp signaling. Dev Dyn 2010; 239:784-97. [PMID: 20112345 DOI: 10.1002/dvdy.22219] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
During dorsal closure in Drosophila, signaling events in the dorsalmost row of epidermal cells (DME cells) direct the migration of lateral epidermal sheets towards the dorsal midline where they fuse to enclose the embryo. A Jun amino-terminal kinase (JNK) cascade in the DME cells induces the expression of Decapentaplegic (Dpp). Dpp signaling then regulates the cytoskeleton in the DME cells and amnioserosa to affect the cell shape changes necessary to complete dorsal closure. We identified a mutation in Sec61alpha that specifically perturbs dorsal closure. Sec61alpha encodes the main subunit of the translocon complex for co-translational import of proteins into the ER. JNK signaling is normal in Sec61alpha mutant embryos, but Dpp signaling is attenuated and the DME cells fail to maintain an actinomyosin cable as epithelial migration fails. Consistent with this model, dorsal closure is rescued in Sec61alpha mutant embryos by an activated form of the Dpp receptor Thick veins.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, USA
| | | |
Collapse
|
29
|
Belacortu Y, Weiss R, Kadener S, Paricio N. Expression of Drosophila Cabut during early embryogenesis, dorsal closure and nervous system development. Gene Expr Patterns 2010; 11:190-201. [PMID: 21109026 DOI: 10.1016/j.gep.2010.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 11/05/2010] [Accepted: 11/18/2010] [Indexed: 01/21/2023]
Abstract
cabut (cbt) encodes a transcription factor involved in Drosophila dorsal closure (DC), and it is expressed in embryonic epithelial sheets and yolk cell during this process upon activation of the Jun N-terminal kinase (JNK) signaling pathway. Additional studies suggest that cbt may have a role in multiple developmental processes. To analyze Cbt localization through embryogenesis, we generated a Cbt specific antibody that has allowed detecting new Cbt expression patterns. Immunohistochemical analyses on syncytial embryos and S2 cells reveal that Cbt is localized on the surface of mitotic chromosomes at all mitotic phases. During DC, Cbt is expressed in the yolk cell, in epidermal cells and in the hindgut, but also in amnioserosal cells, which also contribute to the process, albeit cbt transcripts were not detected in that tissue. At later embryonic stages, Cbt is expressed in neurons and glial cells in the central nervous system, and is detected in axons of the central and peripheral nervous systems. Most of these expression patterns are recapitulated by GFP reporter gene constructs driven by different cbt genomic regions. Moreover, they have been further validated by immunostainings of embryos from other Drosophila species, thus suggesting that Cbt function during embryogenesis appears to be conserved in evolution.
Collapse
Affiliation(s)
- Yaiza Belacortu
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjasot, Spain
| | | | | | | |
Collapse
|
30
|
Seo J, Asaoka Y, Nagai Y, Hirayama J, Yamasaki T, Namae M, Ohata S, Shimizu N, Negishi T, Kitagawa D, Kondoh H, Furutani-Seiki M, Penninger JM, Katada T, Nishina H. Negative regulation of wnt11 expression by Jnk signaling during zebrafish gastrulation. J Cell Biochem 2010; 110:1022-37. [PMID: 20564202 DOI: 10.1002/jcb.22616] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Stress-induced Sapk/Jnk signaling is involved in cell survival and apoptosis. Recent studies have increased our understanding of the physiological roles of Jnk signaling in embryonic development. However, still unclear is the precise function of Jnk signaling during gastrulation, a critical step in the establishment of the vertebrate body plan. Here we use morpholino-mediated knockdown of the zebrafish orthologs of the Jnk activators Mkk4 and Mkk7 to examine the effect of Jnk signaling abrogation on early vertebrate embryogenesis. Depletion of zebrafish Mkk4b led to abnormal convergent extension (CE) during gastrulation, whereas Mkk7 morphants exhibited defective somitogenesis. Surprisingly, Mkk4b morphants displayed marked upregulation of wnt11, which is the triggering ligand of CE and stimulates Jnk activation via the non-canonical Wnt pathway. Conversely, ectopic activation of Jnk signaling by overexpression of an active form of Mkk4b led to wnt11 downregulation. Mosaic lineage tracing studies revealed that Mkk4b-Jnk signaling suppressed wnt11 expression in a non-cell-autonomous manner. These findings provide the first evidence that wnt11 itself is a downstream target of the Jnk cascade in the non-canonical Wnt pathway. Our work demonstrates that Jnk activation is indispensable for multiple steps during vertebrate body plan formation. Furthermore, non-canonical Wnt signaling may coordinate vertebrate CE movements by triggering Jnk activation that represses the expression of the CE-triggering ligand wnt11.
Collapse
Affiliation(s)
- Jungwon Seo
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rousset R, Bono-Lauriol S, Gettings M, Suzanne M, Spéder P, Noselli S. The Drosophila serine protease homologue Scarface regulates JNK signalling in a negative-feedback loop during epithelial morphogenesis. Development 2010; 137:2177-86. [PMID: 20530545 DOI: 10.1242/dev.050781] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In Drosophila melanogaster, dorsal closure is a model of tissue morphogenesis leading to the dorsal migration and sealing of the embryonic ectoderm. The activation of the JNK signal transduction pathway, specifically in the leading edge cells, is essential to this process. In a genome-wide microarray screen, we identified new JNK target genes during dorsal closure. One of them is the gene scarface (scaf), which belongs to the large family of trypsin-like serine proteases. Some proteins of this family, like Scaf, bear an inactive catalytic site, representing a subgroup of serine protease homologues (SPH) whose functions are poorly understood. Here, we show that scaf is a general transcriptional target of the JNK pathway coding for a secreted SPH. scaf loss-of-function induces defects in JNK-controlled morphogenetic events such as embryonic dorsal closure and adult male terminalia rotation. Live imaging of the latter process reveals that, like for dorsal closure, JNK directs the dorsal fusion of two epithelial layers in the pupal genital disc. Genetic data show that scaf loss-of-function mimics JNK over-activity. Moreover, scaf ectopic expression aggravates the effect of the JNK negative regulator puc on male genitalia rotation. We finally demonstrate that scaf acts as an antagonist by negatively regulating JNK activity. Overall, our results identify the SPH-encoding gene scaf as a new transcriptional target of JNK signalling and reveal the first secreted regulator of the JNK pathway acting in a negative-feedback loop during epithelial morphogenesis.
Collapse
Affiliation(s)
- Raphaël Rousset
- University of Nice Sophia-Antipolis, UMR 6543 CNRS, Institute of Developmental Biology and Cancer, Parc Valrose, 06108 Nice CEDEX2, France
| | | | | | | | | | | |
Collapse
|
32
|
Regulation of axonal development by the nuclear protein hindsight (pebbled) in the Drosophila visual system. Dev Biol 2010; 344:911-21. [PMID: 20541542 DOI: 10.1016/j.ydbio.2010.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 11/24/2022]
Abstract
The molecules and networks involved in the process of acquisition and maintenance of the form of a mature neuron are not completely known. Using a misexpression screen we identified the gene hindsight as a gene involved in the process of acquisition of the neuronal morphogenesis in the Drosophila adult nervous system. hindsight encodes a transcription factor known for its role in early developmental processes such as embryonic germ band retraction and dorsal closure, as well as in the establishment of cell morphology, planar cell polarity, and epithelial integrity during retinal development. We describe here a novel function for HNT by showing that both loss and gain of function of HNT affects the pathfinding of the photoreceptors axons. By manipulating the timing and level of HNT expression, together with the number of cells manipulated we show here that the function of HNT in axonal guidance is independent of the HNT functions previously reported in retinal cells. Based on genetic interaction experiments we show that part of HNT function in axonal development is exerted through the regulation of genes involved in the dynamics of the actin cytoskeleton.
Collapse
|
33
|
Szuplewski S, Fraisse-Véron I, George H, Terracol R. vrille is required to ensure tracheal integrity in Drosophila embryo. Dev Growth Differ 2010; 52:409-18. [DOI: 10.1111/j.1440-169x.2010.01186.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Lennox AL, Stronach B. POSH misexpression induces caspase-dependent cell death in Drosophila. Dev Dyn 2010; 239:651-64. [PMID: 20014406 DOI: 10.1002/dvdy.22186] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
POSH (Plenty of SH3 domains) is a scaffold for signaling proteins regulating cell survival. Specifically, POSH promotes assembly of a complex including Rac GTPase, mixed lineage kinase (MLK), MKK7, and Jun kinase (JNK). In Drosophila, genetic analysis implicated POSH in Tak1-dependent innate immune response, in part through regulation of JNK signaling. Homologs of the POSH signaling complex components, MLK and MKK7, are essential in Drosophila embryonic dorsal closure. Using a gain-of-function approach, we tested whether POSH plays a role in this process. Ectopic expression of POSH in the embryo causes dorsal closure defects due to apoptosis of the amnioserosa, but ectodermal JNK signaling is normal. Phenotypic consequences of POSH expression were found to be dependent on Drosophila Nc, the caspase-9 homolog, but only partially on Tak1 and not at all on Slpr and Hep. These results suggest that POSH may use different signaling complexes to promote cell death in distinct contexts.
Collapse
Affiliation(s)
- Ashley L Lennox
- Department of Biological Sciences, 202 Life Sciences Annex, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
35
|
Ghosal K, Naples SP, Rabe AR, Killian KA. Agonistic behavior and electrical stimulation of the antennae induces Fos-like protein expression in the male cricket brain. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2010; 74:38-51. [PMID: 20422717 DOI: 10.1002/arch.20360] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Immediate early genes (IEG) such as c-Fos and Fos-related antigens (FRA) have been used as markers of neuronal activation. In this study, we determined whether the expression of c-Fos/FRAs is increased in the brains of adult male Acheta domesticus crickets following agonistic interactions. We looked for c-Fos/FRA proteins in the brain of un-fought, control male crickets and of dominant and subordinate male crickets sacrificed at different time periods following an agonistic interaction. Using immunoblot analysis, we found four different c-Fos/FRA-like proteins in the adult cricket brain. Continuous agonistic interaction increased c-Fos/FRA protein expression in the brains of subordinate males compared to control and dominant males. In addition, direct electrical stimulation of the male cricket antennae increased c-Fos/FRA-like protein in the brain. We identified the specific brain regions that exhibit c-Fos/FRA-like immunoreactivity in crickets. We detected c-Fos/FRA-like cellular immunoreactivity in different functional regions of the adult brain including the pars intercerebralis, protocerebrum, deutocerebrum, and the cortex of the mushroom bodies.
Collapse
Affiliation(s)
- Kaushik Ghosal
- Department of Zoology and Center for Neuroscience, Miami University, Oxford, Ohio 45056, USA
| | | | | | | |
Collapse
|
36
|
Haghayeghi A, Sarac A, Czerniecki S, Grosshans J, Schöck F. Pellino enhances innate immunity in Drosophila. Mech Dev 2010; 127:301-7. [PMID: 20117206 DOI: 10.1016/j.mod.2010.01.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 01/19/2010] [Accepted: 01/25/2010] [Indexed: 01/02/2023]
Abstract
The innate immune response is a defense mechanism against infectious agents in both vertebrates and invertebrates, and is in part mediated by the Toll pathway. Toll receptor activation upon exposure to bacteria causes stimulation of Pelle/IRAK kinase, eventually resulting in translocation of the transcription factor NF-kappaB to the nucleus. Here we show that Pellino, a highly conserved protein interacting with activated Pelle/IRAK, acts as a positive regulator of innate immunity in Drosophila.
Collapse
Affiliation(s)
- Amirhossein Haghayeghi
- Department of Biology, McGill University, 1205 Dr. Penfield Avenue, Montreal, Que., Canada H3A 1B1
| | | | | | | | | |
Collapse
|
37
|
Panfilio KA, Roth S. Epithelial reorganization events during late extraembryonic development in a hemimetabolous insect. Dev Biol 2010; 340:100-15. [PMID: 20045678 DOI: 10.1016/j.ydbio.2009.12.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 12/18/2009] [Accepted: 12/19/2009] [Indexed: 01/07/2023]
Abstract
As extra-embryonic tissues, the amnion and serosa are not considered to contribute materially to the insect embryo, yet they must execute an array of morphogenetic movements before they are dispensable. In hemimetabolous insects, these movements have been known for over a century, but they have remained virtually unexamined. This study addresses late extraembryonic morphogenesis in the milkweed bug, Oncopeltus fasciatus. Cell shape changes and apoptosis profiles are used to characterize the membranes as they undergo a large repertoire of final reorganizational events that reposition the embryo (katatrepsis), and eliminate the membranes themselves in an ordered fashion (dorsal closure). A number of key features were identified. First, amnion-serosa "fusion" involves localized apoptosis in the amnion and the formation of a supracellular actin purse string at the amnion-serosa border. During katatrepsis, a 'focus' of serosal cells undergoes precocious columnarization and may serve as an anchor for contraction. Lastly, dorsal closure involves novel modifications of the amnion and embryonic flank that are without counterpart during the well-known process of dorsal closure in the fruit fly Drosophila melanogaster. These data also address the long-standing question of the final fate of the amnion: it undergoes apoptosis during dorsal closure and thus is likely to be solely extraembryonic.
Collapse
Affiliation(s)
- Kristen A Panfilio
- Institute for Developmental Biology, University of Cologne, Gyrhofstrabetae 17, 50931 Cologne, Germany.
| | | |
Collapse
|
38
|
Sasikumar S, Roy JK. Developmental expression of Rab11, a small GTP-binding protein inDrosophilaepithelia. Genesis 2009; 47:32-9. [DOI: 10.1002/dvg.20441] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
39
|
Zahedi B, Shen W, Xu X, Chen X, Mahey M, Harden N. Leading edge-secreted Dpp cooperates with ACK-dependent signaling from the amnioserosa to regulate myosin levels during dorsal closure. Dev Dyn 2008; 237:2936-46. [PMID: 18816840 DOI: 10.1002/dvdy.21722] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Dorsal closure of the Drosophila embryo is an epithelial fusion in which the epidermal flanks migrate to close a hole in the epidermis occupied by the amnioserosa, a process driven in part by myosin-dependent cell shape change. Dpp signaling is required for the morphogenesis of both tissues, where it promotes transcription of myosin from the zipper (zip) gene. Drosophila has two members of the activated Cdc42-associated kinase (ACK) family: DACK and PR2. Overexpression of DACK in embryos deficient in Dpp signaling can restore zip expression and suppress dorsal closure defects, while reducing the levels of DACK and PR2 simultaneously using mutations or amnioserosa-specific knock down by RNAi results in loss of zip expression. ACK function in the amnioserosa may generate a signal cooperating with Dpp secreted from the epidermis in driving zip expression in these two tissues, ensuring that cell shape changes in dorsal closure occur in a coordinated manner.
Collapse
Affiliation(s)
- Baharak Zahedi
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
40
|
Regulation of cell adhesion and collective cell migration by hindsight and its human homolog RREB1. Curr Biol 2008; 18:532-7. [PMID: 18394891 DOI: 10.1016/j.cub.2008.03.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 02/04/2008] [Accepted: 03/11/2008] [Indexed: 12/14/2022]
Abstract
Cell movements represent a major driving force in embryonic development, tissue repair, and tumor metastasis [1]. The migration of single cells has been well studied, predominantly in cell culture [2, 3]; however, in vivo, a greater variety of modes of cell movement occur, including the movements of cells in clusters, strands, sheets, and tubes, also known as collective cell migrations [4, 5]. In spite of the relevance of these types of movements in both normal and pathological conditions, the molecular mechanisms that control them remain predominantly unknown. Epithelial follicle cells of the Drosophila ovary undergo several dynamic morphological changes, providing a genetically tractable model [6]. We found that anterior follicle cells, including border cells, mutant for the gene hindsight (hnt) accumulated excess cell-cell adhesion molecules and failed to undergo their normal collective movements. In addition, HNT affected border cell cluster cohesion and motility via effects on the JNK and STAT pathways, respectively. Interestingly, reduction of expression of the mammalian homolog of HNT, RREB1, by siRNA inhibited collective cell migration in a scratch-wound healing assay of MCF10A mammary epithelial cells, suppressed surface activity, retarded cell spreading after plating, and led to the formation of immobile, tightly adherent cell colonies. We propose that HNT and RREB1 are essential to reduce cell-cell adhesion when epithelial cells within an interconnected group undergo dynamic changes in cell shape.
Collapse
|
41
|
Abstract
Drosophila melanogaster is a leading genetic model system in nervous system development and disease research. Using the power of fly genetics in traumatic axonal injury research will significantly speed up the characterization of molecular processes that control axonal regeneration in the CNS. We developed a versatile and physiologically robust preparation for the long-term culture of the whole Drosophila brain. We use this method to develop a novel Drosophila model for CNS axonal injury and regeneration. We first show that, similar to mammalian CNS axons, injured adult wild-type fly CNS axons fail to regenerate, whereas adult-specific enhancement of protein kinase A activity increases the regenerative capacity of lesioned neurons. Combined, these observations suggest conservation of neuronal regeneration mechanisms after injury. We next exploit this model to explore pathways that induce robust regeneration and find that adult-specific activation of c-Jun N-terminal protein kinase signaling is sufficient for de novo CNS axonal regeneration injury, including the growth of new axons past the lesion site and into the normal target area.
Collapse
|
42
|
Vilmos P, Henn L, Szathmári M, Lukácsovich T, Sipos L, Erdélyi M. Application of the dual-tagging gene trap method combined with a novel automatic selection system to identify genes involved in germ cell development in Drosophila melanogaster. ACTA BIOLOGICA HUNGARICA 2008; 58 Suppl:81-94. [PMID: 18297796 DOI: 10.1556/abiol.58.2007.suppl.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The passage of highly specialized germ cells to future generations is essential for the maintenance of species. To date, conventional genetic screens identified relatively few genes that are involved in germ cell development. We aimed to identify germ line specific genes on the X chromosome of Drosophila melanogaster by the application of a new method: the dual-tagging gene-trap system (GT). A modified version of the gene-trap element was used in our experiments and the resulting insertional mutants were screened for grandchild-less phenotype with the help of the attached-X system and a sensitized genetic background developed in our laboratory. Among the 800 insertions mapped to the X chromosome 33 new mutations were identified that exhibited grandchild-less phenotype, 6 gave visible phenotype and 12 were conditional lethal. The cloning of a selected group of the 33 lines showing grandchild-less phenotype confirmed that we have identified new candidates for genes involved in germ cell development. One of them named pebbled (peb) is discussed in details in this paper. Finally, we also describe a novel automatic selection system developed in our laboratory which enables the extension of the GT mutagenesis to the autosomes.
Collapse
Affiliation(s)
- P Vilmos
- Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | | | | | | | | | | |
Collapse
|
43
|
Extraembryonic development in insects and the acrobatics of blastokinesis. Dev Biol 2008; 313:471-91. [DOI: 10.1016/j.ydbio.2007.11.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 10/11/2007] [Accepted: 11/02/2007] [Indexed: 01/04/2023]
|
44
|
Sun J, Deng WM. Hindsight mediates the role of notch in suppressing hedgehog signaling and cell proliferation. Dev Cell 2007; 12:431-42. [PMID: 17336908 PMCID: PMC1851662 DOI: 10.1016/j.devcel.2007.02.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Revised: 12/15/2006] [Accepted: 02/08/2007] [Indexed: 11/28/2022]
Abstract
Temporal and spatial regulation of proliferation and differentiation by signaling pathways is essential for animal development. Drosophila follicular epithelial cells provide an excellent model system for the study of temporal regulation of cell proliferation. In follicle cells, the Notch pathway stops proliferation and promotes a switch from the mitotic cycle to the endocycle. Here, we show that zinc-finger transcription factor Hindsight mediates the role of Notch in regulating cell differentiation and the switch of cell-cycle programs. Hindsight is required and sufficient to stop proliferation and induce the transition to the endocycle. To do so, it represses string, Cut, and Hedgehog signaling, which promote proliferation during early oogenesis. Hindsight, along with another zinc-finger protein, Tramtrack, downregulates Hedgehog signaling through transcriptional repression of cubitus interruptus. Our studies suggest that Hindsight bridges the two antagonistic pathways, Notch and Hedgehog, in the temporal regulation of follicle-cell proliferation and differentiation.
Collapse
|
45
|
Srahna M, Leyssen M, Choi CM, Fradkin LG, Noordermeer JN, Hassan BA. A signaling network for patterning of neuronal connectivity in the Drosophila brain. PLoS Biol 2007; 4:e348. [PMID: 17032066 PMCID: PMC1592317 DOI: 10.1371/journal.pbio.0040348] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Accepted: 08/21/2006] [Indexed: 01/22/2023] Open
Abstract
The precise number and pattern of axonal connections generated during brain development regulates animal behavior. Therefore, understanding how developmental signals interact to regulate axonal extension and retraction to achieve precise neuronal connectivity is a fundamental goal of neurobiology. We investigated this question in the developing adult brain of Drosophila and find that it is regulated by crosstalk between Wnt, fibroblast growth factor (FGF) receptor, and Jun N-terminal kinase (JNK) signaling, but independent of neuronal activity. The Rac1 GTPase integrates a Wnt-Frizzled-Disheveled axon-stabilizing signal and a Branchless (FGF)-Breathless (FGF receptor) axon-retracting signal to modulate JNK activity. JNK activity is necessary and sufficient for axon extension, whereas the antagonistic Wnt and FGF signals act to balance the extension and retraction required for the generation of the precise wiring pattern.
Collapse
Affiliation(s)
- Mohammed Srahna
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, Flanders Interuniversity Institute for Biotechnology (VIB), University of Leuven School of Medicine, Leuven, Belgium
| | - Maarten Leyssen
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, Flanders Interuniversity Institute for Biotechnology (VIB), University of Leuven School of Medicine, Leuven, Belgium
| | - Ching Man Choi
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, Flanders Interuniversity Institute for Biotechnology (VIB), University of Leuven School of Medicine, Leuven, Belgium
| | - Lee G Fradkin
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Jasprina N Noordermeer
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Bassem A Hassan
- Laboratory of Neurogenetics, Department of Molecular and Developmental Genetics, Flanders Interuniversity Institute for Biotechnology (VIB), University of Leuven School of Medicine, Leuven, Belgium
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
46
|
Dow LE, Humbert PO. Polarity Regulators and the Control of Epithelial Architecture, Cell Migration, and Tumorigenesis. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 262:253-302. [PMID: 17631191 DOI: 10.1016/s0074-7696(07)62006-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A large body of work on Drosophila melanogaster has identified and characterized a number of key polarity regulators, many of which are required for the regulation of multiple other processes including proliferation, migration, invasion, and tumorigenesis. Humans possess either single or multiple homologues of each of the Drosophila polarity proteins, and in most cases, these are highly conserved between species, implying an important and conserved function for each of the polarity complexes. Recent studies in cultured mammalian epithelial cells have shed some light on the requirement for the polarity complexes in the regulation of epithelial cell function, including an unexpected link to the regulation of directed cell migration. However, many questions still remain regarding the molecular mechanisms of polarity regulation and whether disruption of polarity protein function is an important step in the development of human cancers. Here we will review what is currently understood about the regulation of cell polarity, migration, and invasion and the level of functional conservation between Drosophila and mammalian tissues. Particular reference will be made as to how the Scribble and Par polarity complexes may be involved in the regulation of apical-basal polarity, migration, and tumorigenesis.
Collapse
Affiliation(s)
- Lukas E Dow
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Center, Melbourne, Australia
| | | |
Collapse
|
47
|
Matsumoto R, Sugimoto M. Dermal matrix proteins initiate re-epithelialization but are not sufficient for coordinated epidermal outgrowth in a new fish skin culture model. Cell Tissue Res 2006; 327:249-65. [PMID: 17043792 DOI: 10.1007/s00441-006-0310-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 07/17/2006] [Indexed: 12/18/2022]
Abstract
We have established a new culture system to study re-epithelialization during fish epidermal wound healing. In this culture system, fetal bovine serum (FBS) stimulates the epidermal outgrowth of multi-cellular layers from scale skin mounted on a coverslip, even when cell proliferation is blocked. The rate of outgrowth is about 0.4 mm/h, and at 3 h after incubation, the area occupied by the epidermal sheet is nine times larger than the area of the original scale skin. Cells at the bottom of the outgrowth show a migratory phenotype with lamellipodia, and "purse string"-like actin bundles have been found over the leading-edge cells with polarized lamellipodia. In the superficial cells, re-development of adherens junctions and microridges has been detected, together with the appearance and translocation of phosphorylated p38 MAPK into nuclear areas. Thus, this culture system provides an excellent model to study the mechanisms of epidermal outgrowth accompanied by migration and re-differentiation. We have also examined the role of extracellular matrix proteins in the outgrowth. Type I collagen or fibronectin stimulates moderate outgrowth in the absence of FBS, but development of microridges and the distribution of phosphorylated p38 MAPK are attenuated in the superficial cells. In addition, the leading-edge cells do not have apparent "purse string"-like actin bundles. The outgrowth stimulated by FBS is inhibited by laminin. These results suggest that dermal substrates such as type I collagen and fibronectin are able to initiate epidermal outgrowth but require other factors to enhance such outgrowth, together with coordinated alterations in cellular phenotype.
Collapse
Affiliation(s)
- Reiko Matsumoto
- Department of Biomolecular Science, Faculty of Science, Toho University, Miyama 2-2-1, Funabashi, Chiba, Japan
| | | |
Collapse
|
48
|
Laplante C, Nilson LA. Differential expression of the adhesion molecule Echinoid drives epithelial morphogenesis in Drosophila. Development 2006; 133:3255-64. [PMID: 16854971 DOI: 10.1242/dev.02492] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Epithelial morphogenesis requires cell movements and cell shape changes coordinated by modulation of the actin cytoskeleton. We identify a role for Echinoid (Ed), an immunoglobulin domain-containing cell-adhesion molecule, in the generation of a contractile actomyosin cable required for epithelial morphogenesis in both the Drosophila ovarian follicular epithelium and embryo. Analysis of ed mutant follicle cell clones indicates that the juxtaposition of wild-type and ed mutant cells is sufficient to trigger actomyosin cable formation. Moreover, in wild-type ovaries and embryos, specific epithelial domains lack detectable Ed, thus creating endogenous interfaces between cells with and without Ed; these interfaces display the same contractile characteristics as the ectopic Ed expression borders generated by ed mutant clones. In the ovary, such an interface lies between the two cell types of the dorsal appendage primordia. In the embryo, Ed is absent from the amnioserosa during dorsal closure, generating an Ed expression border with the lateral epidermis that coincides with the actomyosin cable present at this interface. In both cases, ed mutant epithelia exhibit loss of this contractile structure and subsequent defects in morphogenesis. We propose that local modulation of the cytoskeleton at Ed expression borders may represent a general mechanism for promoting epithelial morphogenesis.
Collapse
Affiliation(s)
- Caroline Laplante
- Department of Biology, McGill University, 1205 Doctor Penfield Avenue, Montréal, QC H3A 1B1, Canada
| | | |
Collapse
|
49
|
Homsy JG, Jasper H, Peralta XG, Wu H, Kiehart DP, Bohmann D. JNK signaling coordinates integrin and actin functions during Drosophila embryogenesis. Dev Dyn 2006; 235:427-34. [PMID: 16317725 DOI: 10.1002/dvdy.20649] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial movements are key morphogenetic events in animal development. They are driven by multiple mechanisms, including signal-dependent changes in cytoskeletal organization and in cell adhesion. Such processes must be controlled precisely and coordinated to accurately sculpt the three-dimensional form of the developing organism. By observing the Drosophila epidermis during embryonic development using confocal time-lapse microscopy, we have investigated how signaling through the Jun-N-terminal kinase (JNK) pathway governs the tissue sheet movements that result in dorsal closure (DC). We find that JNK controls the polymerization of actin into a cable at the epidermal leading edge as previously suggested, as well as the joining (zipping) of the contralateral epithelial cell sheets. Here, we show that zipping is mediated by regulation of the integrins myospheroid and scab. Our data demonstrate that JNK signaling regulates a set of target genes that cooperate to facilitate epithelial movement and closure.
Collapse
Affiliation(s)
- Jason G Homsy
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | |
Collapse
|
50
|
Polgar K, Burrow CR, Hyink DP, Fernandez H, Thornton K, Li X, Gusella GL, Wilson PD. Disruption of polycystin-1 function interferes with branching morphogenesis of the ureteric bud in developing mouse kidneys. Dev Biol 2005; 286:16-30. [PMID: 16122726 DOI: 10.1016/j.ydbio.2005.06.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2004] [Revised: 06/09/2005] [Accepted: 06/30/2005] [Indexed: 12/23/2022]
Abstract
The polycystic kidney disease (PKD1) gene-encoded protein, polycystin-1, is developmentally regulated, with highest expression levels seen in normal developing kidneys, where it is distributed in a punctate pattern at the basal surface of ureteric bud epithelia. Overexpression in ureteric epithelial cell membranes of an inhibitory pMyr-GFP-PKD1 fusion protein via a retroviral (VVC) delivery system and microinjection into the ureteric bud lumen of embryonic day 11 mouse metanephric kidneys resulted in disrupted branching morphogenesis. Using confocal quantitative analysis, significant reductions were measured in the numbers of ureteric bud branch points and tips, as well as in the total ureteric bud length, volume and area, while significant increases were seen as dilations of the terminal branches, where significant increases in outer diameter and volumes were measured. Microinjection of an activating 5TM-GFP-PKD1 fusion protein had an opposite effect and showed significant increases in ureteric bud length and area. These are the first studies to experimentally manipulate polycystin-1 expression by transduction in the embryonic mouse kidney and suggest that polycystin-1 plays a critical role in the regulation of epithelial morphogenesis during renal development.
Collapse
Affiliation(s)
- Katalin Polgar
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|