Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38323935

ABSTRACT

Robust linkage between adherens junctions and the actomyosin cytoskeleton allows cells to change shape and move during morphogenesis without tearing tissues apart. The Drosophila multidomain protein Canoe and its mammalian homolog afadin are crucial for this, as in their absence many events of morphogenesis fail. To define the mechanism of action for Canoe, we are taking it apart. Canoe has five folded protein domains and a long intrinsically disordered region. The largest is the Dilute domain, which is shared by Canoe and myosin V. To define the roles of this domain in Canoe, we combined biochemical, genetic and cell biological assays. AlphaFold was used to predict its structure, providing similarities and contrasts with Myosin V. Biochemical data suggested one potential shared function - the ability to dimerize. We generated Canoe mutants with the Dilute domain deleted (CnoΔDIL). Surprisingly, they were viable and fertile. CnoΔDIL localized to adherens junctions and was enriched at junctions under tension. However, when its dose was reduced, CnoΔDIL did not provide fully wild-type function. Furthermore, canoeΔDIL mutants had defects in the orchestrated cell rearrangements of eye development. This reveals the robustness of junction-cytoskeletal connections during morphogenesis and highlights the power of natural selection to maintain protein structure.


Subject(s)
Drosophila Proteins , Myosin Type V , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Myosin Type V/metabolism , Cytoskeleton/metabolism , Intercellular Junctions/metabolism , Adherens Junctions/metabolism , Morphogenesis , Cadherins/metabolism , Mammals/metabolism
2.
bioRxiv ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37905001

ABSTRACT

Robust linkage between cell-cell adherens junctions and the actomyosin cytoskeleton allows cells to change shape and move during morphogenesis without tearing tissues apart. The multidomain protein Drosophila Canoe and its mammalian homolog Afadin are critical for this linkage, and in their absence many events of morphogenesis fail. To define underlying mechanisms, we are taking Canoe apart, using Drosophila as our model. Canoe and Afadin share five folded protein domains, followed by a large intrinsically disordered region. The largest of these folded domains is the Dilute domain, which is found in Canoe/Afadin, their paralogs, and members of the MyosinV family. To define the roles of Canoe's Dilute domain we have combined biochemical, genetic and cell biological assays. Use of the AlphaFold tools revealed the predicted structure of the Canoe/Afadin Dilute domain, providing similarities and contrasts with that of MyosinV. Our biochemical data suggest one potential shared function: the ability to dimerize. We next generated Drosophila mutants with the Dilute domain cleanly deleted. Surprisingly, these mutants are viable and fertile, and CanoeΔDIL protein localizes to adherens junctions and is enriched at junctions under tension. However, when we reduce the dose of CanoeΔDIL protein in a sensitized assay, it becomes clear it does not provide full wildtype function. Further, canoeΔDIL mutants have defects in pupal eye development, another process that requires orchestrated cell rearrangements. Together, these data reveal the robustness in AJ-cytoskeletal connections during multiple embryonic and postembryonic events, and the power of natural selection to maintain protein structure even in robust systems.

3.
J Cell Biol ; 220(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34762121

ABSTRACT

Embryogenesis requires cells to change shape and move without disrupting epithelial integrity. This requires robust, responsive linkage between adherens junctions and the actomyosin cytoskeleton. Using Drosophila morphogenesis, we define molecular mechanisms mediating junction-cytoskeletal linkage and explore the role of mechanosensing. We focus on the junction-cytoskeletal linker Canoe, a multidomain protein. We engineered the canoe locus to define how its domains mediate its mechanism of action. To our surprise, the PDZ and FAB domains, which we thought connected junctions and F-actin, are not required for viability or mechanosensitive recruitment to junctions under tension. The FAB domain stabilizes junctions experiencing elevated force, but in its absence, most cells recover, suggesting redundant interactions. In contrast, the Rap1-binding RA domains are critical for all Cno functions and enrichment at junctions under tension. This supports a model in which junctional robustness derives from a large protein network assembled via multivalent interactions, with proteins at network nodes and some node connections more critical than others.


Subject(s)
Adherens Junctions/metabolism , Cytoskeleton/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Morphogenesis , Alleles , Animals , Cell Survival , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Epithelium/metabolism , Loss of Function Mutation/genetics , Protein Domains
4.
Dev Biol ; 478: 173-182, 2021 10.
Article in English | MEDLINE | ID: mdl-34245727

ABSTRACT

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


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila/growth & development , Photoreceptor Cells, Invertebrate/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Compound Eye, Arthropod/cytology , Computer Simulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Larva/growth & development , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Invertebrate/cytology , Pupa/growth & development , Receptors, Invertebrate Peptide/metabolism , Signal Transduction
5.
G3 (Bethesda) ; 11(1)2021 01 18.
Article in English | MEDLINE | ID: mdl-33561221

ABSTRACT

Tissue function is dependent on correct cellular organization and behavior. As a result, the identification and study of genes that contribute to tissue morphogenesis is of paramount importance to the fields of cell and developmental biology. Many of the genes required for tissue patterning and organization are highly conserved between phyla. This has led to the emergence of several model organisms and developmental systems that are used to study tissue morphogenesis. One such model is the Drosophila melanogaster pupal eye that has a highly stereotyped arrangement of cells. In addition, the pupal eye is postmitotic that allows for the study of tissue morphogenesis independent from any effects of proliferation. While the changes in cell morphology and organization that occur throughout pupal eye development are well documented, less is known about the corresponding transcriptional changes that choreograph these processes. To identify these transcriptional changes, we dissected wild-type Canton S pupal eyes and performed RNA-sequencing. Our analyses identified differential expression of many loci that are documented regulators of pupal eye morphogenesis and contribute to multiple biological processes including signaling, axon projection, adhesion, and cell survival. We also identified differential expression of genes not previously implicated in pupal eye morphogenesis such as components of the Toll pathway, several non-classical cadherins, and components of the muscle sarcomere, which could suggest these loci function as novel patterning factors.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Morphogenesis , Pupa
6.
Dev Biol ; 464(1): 53-70, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32464117

ABSTRACT

Hippo signaling is an important regulator of tissue size, but it also has a lesser-known role in tissue morphogenesis. Here we use the Drosophila pupal eye to explore the role of the Hippo effector Yki and its cofactor Mask in morphogenesis. We found that Mask is required for the correct distribution and accumulation of adherens junctions and appropriate organization of the cytoskeleton. Accordingly, disrupting mask expression led to severe mis-patterning and similar defects were observed when yki was reduced or in response to ectopic wts. Further, the patterning defects generated by reducing mask expression were modified by Hippo pathway activity. RNA-sequencing revealed a requirement for Mask for appropriate expression of numerous genes during eye morphogenesis. These included genes implicated in cell adhesion and cytoskeletal organization, a comprehensive set of genes that promote cell survival, and numerous signal transduction genes. To validate our transcriptome analyses, we then considered two loci that were modified by Mask activity: FER and Vinc, which have established roles in regulating adhesion. Modulating the expression of either locus modified mask mis-patterning and adhesion phenotypes. Further, expression of FER and Vinc was modified by Yki. It is well-established that the Hippo pathway is responsive to changes in cell adhesion and the cytoskeleton, but our data indicate that Hippo signaling also regulates these structures.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Eye/embryology , Gene Expression Regulation, Developmental/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Organogenesis/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Eye/cytology , Eye Proteins/genetics , Eye Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins
7.
J Am Soc Nephrol ; 30(7): 1220-1237, 2019 07.
Article in English | MEDLINE | ID: mdl-31235616

ABSTRACT

BACKGROUND: CD2-associated protein (CD2AP), a slit diaphragm-associated scaffolding protein involved in survival and regulation of the cytoskeleton in podocytes, is considered a "stabilizer" of the slit diaphragm complex that connects the slit diaphragm protein nephrin to the cytoskeleton of the cell. Tyrosine phosphorylation of slit diaphragm molecules can influence their surface expression, but it is unknown whether tyrosine phosphorylation events of CD2AP are also physiologically relevant to slit diaphragm stability. METHODS: We used isoelectric focusing, western blot analysis, and immunofluorescence to investigate phosphorylation of CD2AP, and phospho-CD2AP antibodies and site-directed mutagenesis to define the specific phosphorylated tyrosine residues. We used cross-species rescue experiments in Cd2apKD zebrafish and in Drosophila cindrRNAi mutants to define the physiologic relevance of CD2AP phosphorylation of the tyrosine residues. RESULTS: We found that VEGF-A stimulation can induce a tyrosine phosphorylation response in CD2AP in podocytes, and that these phosphorylation events have an important effect on slit diaphragm protein localization and functionality in vivo. We demonstrated that tyrosine in position Y10 of the SH3-1 domain of CD2AP is indispensable for CD2AP function in vivo. We found that the binding affinity of nephrin to CD2AP is significantly enhanced in the absence of Y10; however, unexpectedly, this increased affinity leads not to stabilization but to functional impairment of the glomerular filtration barrier. CONCLUSIONS: Our findings provide insight into CD2AP and its phosphorylation in the context of slit diaphragm functionality, and indicate a fine-tuned affinity balance of CD2AP and nephrin that is influenced by receptor tyrosine kinase stimulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/chemistry , Tyrosine/metabolism , Animals , Drosophila melanogaster , HEK293 Cells , Humans , Membrane Proteins/metabolism , Phosphorylation , Podocytes/metabolism , Protein Stability , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish
8.
J Vis Exp ; (145)2019 03 15.
Article in English | MEDLINE | ID: mdl-30933080

ABSTRACT

The Drosophila pupal retina provides an excellent model system for the study of morphogenetic processes during development. In this paper, we present a reliable protocol for the dissection of the delicate Drosophila pupal retina. Our surgical approach utilizes readily-available microdissection tools to open pupae and precisely extract eye-brain complexes. These can be fixed, subjected to immunohistochemistry, and retinas then mounted onto microscope slides and imaged if the goal is to detect cellular or subcellular structures. Alternatively, unfixed retinas can be isolated from brain tissue, lysed in appropriate buffers and utilized for protein gel electrophoresis or mRNA extraction (to assess protein or gene expression, respectively). Significant practice and patience may be required to master the microdissection protocol described, but once mastered, the protocol enables relatively quick isolation of mainly undamaged retinas.


Subject(s)
Dissection , Drosophila melanogaster/cytology , RNA/isolation & purification , Retina/cytology , Retina/surgery , Animals , Blotting, Western , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Immunohistochemistry , Pupa/cytology , RNA/metabolism , Retina/metabolism
9.
Dev Biol ; 433(1): 94-107, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29133184

ABSTRACT

Apoptosis is crucial during the morphogenesis of most organs and tissues, and is utilized for tissues to achieve their proper size, shape and patterning. Many signaling pathways contribute to the precise regulation of apoptosis. Here we show that Jun N-terminal Kinase (JNK) activity contributes to the coordinated removal of interommatidial cells via apoptosis in the Drosophila pupal retina. This is consistent with previous findings that JNK activity promotes apoptosis in other epithelia. However, we found that JNK activity is repressed by Cindr (the CIN85 and CD2AP ortholog) in order to promote cell survival. Reducing the amount of Cindr resulted in ectopic cell death. Increased expression of the Drosophila JNK basket in the setting of reduced cindr expression was found to result in even more severe apoptosis, whilst ectopic death was found to be reduced if retinas were heterozygous for basket. Hence Cindr is required to properly restrict JNK-mediated apoptosis in the pupal eye, resulting in the correct number of interommatidial cells. A lack of precise control over developmental apoptosis can lead to improper tissue morphogenesis.


Subject(s)
JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Apoptosis/physiology , Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelium/enzymology , Epithelium/metabolism , Gene Expression Regulation, Developmental/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Microfilament Proteins/metabolism , Morphogenesis , Pupa/metabolism , Retina/cytology , Retina/enzymology , Retina/metabolism , Signal Transduction
10.
PLoS One ; 12(11): e0187571, 2017.
Article in English | MEDLINE | ID: mdl-29117266

ABSTRACT

Ubiquitination is a crucial post-translational modification that can target proteins for degradation. The E3 ubiquitin ligases are responsible for recognizing substrate proteins for ubiquitination, hence providing specificity to the process of protein degradation. Here, we describe a genetic modifier screen that identified E3 ligases that modified the rough-eye phenotype generated by expression of cindrRNAi transgenes during Drosophila eye development. In total, we identified 36 E3 ligases, as well as 4 Cullins, that modified the mild cindrRNA mis-patterning phenotype. This indicates possible roles for these E3s/Cullins in processes that require Cindr function, including cytoskeletal regulation, cell adhesion, cell signaling and cell survival. Three E3 ligases identified in our screen had previously been linked to regulating JNK signaling.


Subject(s)
Body Patterning/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Eye/embryology , Eye/metabolism , Microfilament Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Alleles , Animals , Cullin Proteins/metabolism , Epistasis, Genetic , Genetic Loci , MAP Kinase Signaling System , Phylogeny , Pupil
11.
J Biol Chem ; 291(47): 24406-24417, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27707879

ABSTRACT

MAGI-1 is a multidomain cytosolic scaffolding protein that in the kidney is specifically located at the podocyte slit diaphragm, a specialized junction that is universally injured in proteinuric diseases. There it interacts with several essential molecules, including nephrin and neph1, which are required for slit diaphragm formation and as an intracellular signaling hub. Here, we show that diminished MAGI-1 expression in cultured podocytes reduced nephrin and neph1 membrane localization and weakened tight junction integrity. Global magi1 knock-out mice, however, demonstrated normal glomerular histology and function into adulthood. We hypothesized that a second mild but complementary genetic insult might induce glomerular disease susceptibility in these mice. To identify such a gene, we utilized the developing fly eye to test for functional complementation between MAGI and its binding partners. In this way, we identified diminished expression of fly Hibris (nephrin) or Roughest (neph1) as dramatically exacerbating the effects of MAGI depletion. Indeed, when these combinations were studied in mice, the addition of nephrin, but not neph1, heterozygosity to homozygous deletion of MAGI-1 resulted in spontaneous glomerulosclerosis. In cultured podocytes, MAGI-1 depletion reduced intercellular contact-induced Rap1 activation, a pathway critical for proper podocyte function. Similarly, magi1 knock-out mice showed diminished glomerular Rap1 activation, an effect dramatically enhanced by concomitant nephrin haploinsufficiency. Finally, combined overexpression of MAGI-1 and nephrin increased Rap1 activation, but not when substituting a mutant MAGI-1 that cannot bind nephrin. We conclude that the interaction between nephrin and MAGI-1 regulates Rap1 activation in podocytes to maintain long term slit diaphragm structure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion Molecules , Enzyme Activation , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Guanylate Kinases , Membrane Proteins/genetics , Mice , Mice, Knockout , rap1 GTP-Binding Proteins/genetics
12.
Dev Biol ; 410(2): 135-149, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26772997

ABSTRACT

Epithelia are essential barrier tissues that must be appropriately maintained for their correct function. To achieve this a plethora of protein interactions regulate epithelial cell number, structure and adhesion, and differentiation. Here we show that Cindr (the Drosophila Cin85 and Cd2ap ortholog) is required to maintain epithelial integrity. Reducing Cindr triggered cell delamination and movement. Most delaminating cells died. These behaviors were consistent with JNK activation previously associated with loss of epithelial integrity in response to ectopic oncogene activity. We confirmed a novel interaction between Cindr and Drosophila JNK (dJNK), which when perturbed caused inappropriate JNK signaling. Genetically reducing JNK signaling activity suppressed the effects of reducing Cindr. Furthermore, ectopic JNK signaling phenocopied loss of Cindr and was partially rescued by concomitant cindr over-expression. Thus, correct Cindr-dJNK stoichiometry is essential to maintain epithelial integrity and disturbing this balance may contribute to the pathogenesis of disease states, including cancer.


Subject(s)
Drosophila Proteins/physiology , MAP Kinase Kinase 4/metabolism , Microfilament Proteins/physiology , Animals , Drosophila/genetics , Epithelium/metabolism , Female , Male
13.
Mol Cell Endocrinol ; 413: 145-56, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26112185

ABSTRACT

G-protein coupled receptors (GPCRs) and their ligands provide precise tissue regulation and are therefore often restricted to specific animal phyla. For example, the gonadotropins and their receptors are crucial for vertebrate reproduction but absent from invertebrates. In mammals, LHR mainly couples to the PKA signaling pathway, and CREB is the major transcription factor of this pathway. Here we present the results of expressing elements of the human gonadotropin system in Drosophila. Specifically, we generated transgenic Drosophila expressing the human LH/CG receptor (denoted as LHR), a constitutively active form of LHR, and an hCG analog. We demonstrate activation-dependent signaling by LHR to direct Drosophila phenotypes including lethality and specific midline defects; these phenotypes were due to LHR activation of PKA/CREB pathway activity. That the LHR can act in an invertebrate demonstrates the conservation of factors required for GPCR function among phylogenetically distant organisms. This novel gonadotropin model may assist the identification of new modulators of mammalian fertility by exploiting the powerful genetic and pharmacological tools available in Drosophila.


Subject(s)
Receptors, LH/metabolism , Signal Transduction , Animals , Animals, Genetically Modified , Drosophila melanogaster , Humans , Receptors, LH/genetics
14.
J Vis Exp ; (95): 52120, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25651413

ABSTRACT

Inherent processes of Drosophila pupal development can shift and distort the eye epithelium in ways that make individual cell behavior difficult to track during live cell imaging. These processes include: retinal rotation, cell growth and organismal movement. Additionally, irregularities in the topology of the epithelium, including subtle bumps and folds often introduced as the pupa is prepared for imaging, make it challenging to acquire in-focus images of more than a few ommatidia in a single focal plane. The workflow outlined here remedies these issues, allowing easy analysis of cellular processes during Drosophila pupal eye development. Appropriately-staged pupae are arranged in an imaging rig that can be easily assembled in most laboratories. Ubiquitin-DE-Cadherin:GFP and GMR-GAL4-driven UAS-α-catenin:GFP are used to visualize cell boundaries in the eye epithelium (1-3). After deconvolution is applied to fluorescent images captured at multiple focal planes, maximum projection images are generated for each time point and enhanced using image editing software. Alignment algorithms are used to quickly stabilize superfluous motion, making individual cell behavior easier to track.


Subject(s)
Drosophila/cytology , Retina/cytology , Animals , Animals, Genetically Modified , Drosophila Proteins/analysis , Epithelium , Female , Green Fluorescent Proteins/analysis , Male , Pupa/cytology
15.
Dev Dyn ; 241(12): 1933-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23027549

ABSTRACT

BACKGROUND: Morphogenetic modeling of tissues requires coordinated regulation of adhesion. For its correct patterning, the Drosophila pupal eye requires several Immunoglobulin superfamily cell adhesion molecules (IgCAMs) and the adaptor protein Cindr. Orthologs of these proteins are essential components of specialized junctions of the vertebrate kidney; the Cindr ortholog Cd2ap is essential for the integrity of this structure. RESULTS: Reducing Cindr during fly eye development led to incorrect distribution of the IgCAMs Roughest (Rst) and Hibris (Hbs). Both bound Cindr. Disrupting endocytosis similarly led to Rst and Hbs mis-localization; our data suggests an additional early requirement for endocytosis in regulating Hbs localization or stability. Finally, Rst and Hbs localized correctly only when in stable membrane complexes and we propose that Cindr anchors these to the cytoskeleton. This regulation likely does not extend to IgCAMs Kin of irre (Kirre) and Sticks and stones (Sns) in the pupal eye; neither interacted with Cindr in in vitro assays. Nonetheless, Kirre and Sns partially mis-localized when Cindr was reduced, possibly due to interactions with Rst/Hbs. CONCLUSIONS: Our data suggests Cindr recapitulates both proposed functions of its mammalian orthologs Cd2ap and Cin85: targeting the IgCAMs Rst and Hbs for endocytosis and stabilizing these heterophilic IgCAM complexes.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Endocytosis/physiology , Eye Proteins/genetics , Membrane Proteins/genetics , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism
16.
Mol Biol Cell ; 22(23): 4513-26, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21976699

ABSTRACT

Patterning of the Drosophila pupal eye is characterized by precise cell movements. In this paper, we demonstrate that these movements require an Arf regulatory cycle that connects surface receptors to actin-based movement. dArf6 activity-regulated by the Arf GTPase-activating proteins (ArfGAPs) dAsap and dArfGAP3 and the Arf GTP exchange factors Schizo and dPsd-promoted large cellular extensions; time-lapse microscopy indicated that these extensions presage cell rearrangements into correct epithelial niches. During this process, the Drosophila eye also requires interactions between surface Neph1/nephrin adhesion receptors Roughest and Hibris, which bind the adaptor protein Cindr (CD2AP). We provide evidence that Cindr forms a physical complex with dArfGAP3 and dAsap. Our data suggest this interaction sequesters ArfGAP function to liberate active dArf6 elsewhere in the cell. We propose that a Neph1/nephrin-Cindr/ArfGAP complex accumulates to limit local Arf6 activity and stabilize adherens junctions. Our model therefore links surface adhesion via an Arf6 regulatory cascade to dynamic modeling of the cytoskeleton, accounting for precise cell movements that organize the functional retinal field. Further, we demonstrate a similar relationship between the mammalian Cindr orthologue CD2AP and Arf6 activity in cell motility assays. We propose that this Cindr/CD2AP-mediated regulation of Arf6 is a widely used mechanism in emerging epithelia.


Subject(s)
Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila/growth & development , Epithelium/growth & development , Eye/cytology , Microfilament Proteins/metabolism , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Drosophila Proteins/genetics , Eye/growth & development , Eye Proteins/genetics , Eye Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pupa
17.
PLoS Comput Biol ; 6: e1000841, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20617161

ABSTRACT

We present a computer simulation and associated experimental validation of assembly of glial-like support cells into the interweaving hexagonal lattice that spans the Drosophila pupal eye. This process of cell movements organizes the ommatidial array into a functional pattern. Unlike earlier simulations that focused on the arrangements of cells within individual ommatidia, here we examine the local movements that lead to large-scale organization of the emerging eye field. Simulations based on our experimental observations of cell adhesion, cell death, and cell movement successfully patterned a tracing of an emerging wild-type pupal eye. Surprisingly, altering cell adhesion had only a mild effect on patterning, contradicting our previous hypothesis that the patterning was primarily the result of preferential adhesion between IRM-class surface proteins. Instead, our simulations highlighted the importance of programmed cell death (PCD) as well as a previously unappreciated variable: the expansion of cells' apical surface areas, which promoted rearrangement of neighboring cells. We tested this prediction experimentally by preventing expansion in the apical area of individual cells: patterning was disrupted in a manner predicted by our simulations. Our work demonstrates the value of combining computer simulation with in vivo experiments to uncover novel mechanisms that are perpetuated throughout the eye field. It also demonstrates the utility of the Glazier-Graner-Hogeweg model (GGH) for modeling the links between local cellular interactions and emergent properties of developing epithelia as well as predicting unanticipated results in vivo.


Subject(s)
Compound Eye, Arthropod , Computer Simulation , Drosophila/growth & development , Morphogenesis/physiology , Pupa/growth & development , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Adhesion , Cell Movement , Cell Proliferation , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/ultrastructure , Microscopy, Electron, Transmission , Models, Biological , Mutation , Surface Properties
18.
Fly (Austin) ; 4(3): 182-93, 2010.
Article in English | MEDLINE | ID: mdl-20495361

ABSTRACT

Here we investigate the structural and functional basis of the interactions between Notch and Wingless signalling in Drosophila. Using yeast-two-hybrid and pull-down assays we show that Notch can bind directly a form of Dishevelled that is stabilized upon Wingless signalling. Moreover, we show that the mechanism by which Wingless signalling is able to downregulate Notch is by promoting its ligand-independent traffic to a compartment where it is degraded and that this activity depends on Dishevelled.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Phosphoproteins/physiology , Receptors, Notch/metabolism , Wnt1 Protein/physiology , Animals , Dishevelled Proteins , Drosophila , Female , Ligands , Male , Protein Transport/physiology , Signal Transduction , Two-Hybrid System Techniques
19.
PLoS One ; 4(9): e7008, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19753121

ABSTRACT

BACKGROUND: The Drosophila pupal eye has become a popular paradigm for understanding morphogenesis and tissue patterning. Correct rearrangement of cells between ommatidia is required to organize the ommatidial array across the eye field. This requires cell movement, cell death, changes to cell-cell adhesion, signaling and fate specification. METHODOLOGY: We describe a method to quantitatively assess mis-patterning of the Drosophila pupal eye and objectively calculate a 'mis-patterning score' characteristic of a specific genotype. This entails step-by-step scoring of specific traits observed in pupal eyes dissected 40-42 hours after puparium formation and subsequent statistical analysis of this data. SIGNIFICANCE: This method provides an unbiased quantitative score of mis-patterning severity that can be used to compare the impact of different genetic mutations on tissue patterning.


Subject(s)
Body Patterning , Drosophila/embryology , Drosophila/physiology , Gene Expression Regulation, Developmental , Photoreceptor Cells, Invertebrate/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules, Neuronal/metabolism , Cell Lineage , Data Interpretation, Statistical , Genotype , Microscopy/methods , Mutation , Phenotype , Pupa , Signal Transduction
20.
Dev Biol ; 318(1): 1-16, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18423436

ABSTRACT

Correct cellular patterning is central to tissue morphogenesis, but the role of epithelial junctions in this process is not well-understood. The Drosophila pupal eye provides a sensitive and accessible model for testing the role of junction-associated proteins in cells that undergo dynamic and coordinated movements during development. Mutations in polychaetoid (pyd), the Drosophila homologue of Zonula Occludens-1, are characterized by two phenotypes visible in the adult fly: increased sensory bristle number and the formation of a rough eye produced by poorly arranged ommatidia. We found that Pyd was localized to the adherens junction in cells of the developing pupal retina. Reducing Pyd function in the pupal eye resulted in mis-patterning of the interommatidial cells and a failure to consistently switch cone cell contacts from an anterior-posterior to an equatorial-polar orientation. Levels of Roughest, DE-Cadherin and several other adherens junction-associated proteins were increased at the membrane when Pyd protein was reduced. Further, both over-expression and mutations in several junction-associated proteins greatly enhanced the patterning defects caused by reduction of Pyd. Our results suggest that Pyd modulates adherens junction strength and Roughest-mediated preferential cell adhesion.


Subject(s)
Adherens Junctions/metabolism , Body Patterning , Cell Adhesion/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Pupa/anatomy & histology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Membrane Proteins/genetics , Morphogenesis , Phosphoproteins/genetics , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/embryology , Pupa/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/cytology , Retina/embryology , Signal Transduction/physiology , Tight Junction Proteins , Wings, Animal/anatomy & histology , Wings, Animal/embryology , Zonula Occludens-1 Protein , alpha Catenin/genetics , alpha Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...