Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 452
Filter
1.
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
2.
PLoS Genet ; 17(6): e1009613, 2021 06.
Article in English | MEDLINE | ID: mdl-34161320

ABSTRACT

Color vision in Drosophila melanogaster is based on the expression of five different color-sensing Rhodopsin proteins in distinct subtypes of photoreceptor neurons. Promoter regions of less than 300 base pairs are sufficient to reproduce the unique, photoreceptor subtype-specific rhodopsin expression patterns. The underlying cis-regulatory logic remains poorly understood, but it has been proposed that the rhodopsin promoters have a bipartite structure: the distal promoter region directs the highly restricted expression in a specific photoreceptor subtype, while the proximal core promoter region provides general activation in all photoreceptors. Here, we investigate whether the rhodopsin promoters exhibit a strict specialization of their distal (subtype specificity) and proximal (general activation) promoter regions, or if both promoter regions contribute to generating the photoreceptor subtype-specific expression pattern. To distinguish between these two models, we analyze the expression patterns of a set of hybrid promoters that combine the distal promoter region of one rhodopsin with the proximal core promoter region of another rhodopsin. We find that the function of the proximal core promoter regions extends beyond providing general activation: these regions play a previously underappreciated role in generating the non-overlapping expression patterns of the different rhodopsins. Therefore, cis-regulatory motifs in both the distal and the proximal core promoter regions recruit transcription factors that generate the unique rhodopsin patterns in a combinatorial manner. We compare this combinatorial regulatory logic to the regulatory logic of olfactory receptor genes and discuss potential implications for the evolution of rhodopsins.


Subject(s)
Color Vision/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Photoreceptor Cells, Invertebrate/metabolism , Promoter Regions, Genetic , Rhodopsin/genetics , Transcription Factors/genetics , Animals , Base Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Evolution, Molecular , Female , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Photoreceptor Cells, Invertebrate/classification , Photoreceptor Cells, Invertebrate/cytology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rhodopsin/metabolism , Transcription Factors/metabolism
3.
Arthropod Struct Dev ; 60: 101007, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33341370

ABSTRACT

As an obligate ectoparasite of bats, the bat fly Trichobius frequens (Diptera: Streblidae) inhabits the same subterranean environment as their nocturnal bat hosts. In this study, we characterize the macromorphology, optical architecture, rhabdom anatomy, photoreceptor absorbance, and opsin expression of the significantly reduced visual system in T. frequens resulting from evolution in the dark. The eyes develop over a 21-22 day pupal developmental period, with pigmentation appearing on pupal day 11. After eclosion as an adult, T. frequens eyes consist of on average 8 facets, each overlying a fused rhabdom consisting of anywhere from 11 to 18 estimated retinula cells. The dimensions of the facets and fused rhabdoms are similar to those measured in other nocturnal insects. T. frequens eyes are functional as shown by expression of a Rh1 opsin forming a visual pigment with a peak sensitivity to 487 nm, similar to other dipteran Rh1 opsins. Future studies will evaluate how individuals with such reduced capabilities for spatial vision as well as sensitivity still capture enough visual information to use flight to maneuver through dark habitats.


Subject(s)
Compound Eye, Arthropod/anatomy & histology , Diptera/anatomy & histology , Photoreceptor Cells, Invertebrate/cytology , Animals , Chiroptera/parasitology , Compound Eye, Arthropod/ultrastructure , Diptera/genetics , Diptera/ultrastructure , Female , Gene Expression , Host-Parasite Interactions , Insect Proteins/genetics , Insect Proteins/metabolism , Male , Microscopy, Confocal , Microscopy, Electron, Scanning , Opsins/genetics , Opsins/metabolism , Photoreceptor Cells, Invertebrate/ultrastructure
4.
Dev Biol ; 471: 89-96, 2021 03.
Article in English | MEDLINE | ID: mdl-33333066

ABSTRACT

Diversification of neuronal subtypes often requires stochastic gene regulatory mechanisms. How stochastically expressed transcription factors interact with other regulators in gene networks to specify cell fates is poorly understood. The random mosaic of color-detecting R7 photoreceptor subtypes in Drosophila is controlled by the stochastic on/off expression of the transcription factor Spineless (Ss). In SsON R7s, Ss induces expression of Rhodopsin 4 (Rh4), whereas in SsOFF R7s, the absence of Ss allows expression of Rhodopsin 3 (Rh3). Here, we find that the transcription factor Runt, which is initially expressed in all R7s, is sufficient to promote stochastic Ss expression. Later, as R7s develop, Ss negatively feeds back onto Runt to prevent repression of Rh4 and ensure proper fate specification. Together, stereotyped and stochastic regulatory inputs are integrated into feedforward and feedback mechanisms to control cell fate.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Photoreceptor Cells, Invertebrate/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Rhodopsin/biosynthesis , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Photoreceptor Cells, Invertebrate/cytology , Receptors, Aryl Hydrocarbon/genetics , Rhodopsin/genetics
5.
J Cell Biol ; 219(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33048164

ABSTRACT

Apical domains of epithelial cells often undergo dramatic changes during morphogenesis to form specialized structures, such as microvilli. Here, we addressed the role of lipids during morphogenesis of the rhabdomere, the microvilli-based photosensitive organelle of Drosophila photoreceptor cells. Shotgun lipidomics analysis performed on mutant alleles of the polarity regulator crumbs, exhibiting varying rhabdomeric growth defects, revealed a correlation between increased abundance of hydroxylated sphingolipids and abnormal rhabdomeric growth. This could be attributed to an up-regulation of fatty acid hydroxylase transcription. Indeed, direct genetic perturbation of the hydroxylated sphingolipid metabolism modulated rhabdomere growth in a crumbs mutant background. One of the pathways targeted by sphingolipid metabolism turned out to be the secretory route of newly synthesized Rhodopsin, a major rhabdomeric protein. In particular, altered biosynthesis of hydroxylated sphingolipids impaired apical trafficking via Rab11, and thus apical membrane growth. The intersection of lipid metabolic pathways with apical domain growth provides a new facet to our understanding of apical growth during morphogenesis.


Subject(s)
Morphogenesis , Photoreceptor Cells, Invertebrate/metabolism , Sphingolipids/biosynthesis , Animals , Drosophila Proteins/economics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Hydroxylation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/cytology , Sphingolipids/genetics , rab GTP-Binding Proteins/economics , rab GTP-Binding Proteins/metabolism
6.
PLoS One ; 15(10): e0240451, 2020.
Article in English | MEDLINE | ID: mdl-33052948

ABSTRACT

Cell differentiation and cell fate determination in sensory systems are essential for stimulus discrimination and coding of environmental stimuli. Color vision is based on the differential color sensitivity of retinal photoreceptors, however the developmental programs that control photoreceptor cell differentiation and specify color sensitivity are poorly understood. In Drosophila melanogaster, there is evidence that the color sensitivity of different photoreceptors in the compound eye is regulated by inductive signals between cells, but the exact nature of these signals and how they are propagated remains unknown. We conducted a genetic screen to identify additional regulators of this process and identified a novel mutation in the hibris gene, which encodes an irre cell recognition module protein (IRM). These immunoglobulin super family cell adhesion molecules include human KIRREL and nephrin (NPHS1). hibris is expressed dynamically in the developing Drosophila melanogaster eye and loss-of-function mutations give rise to a diverse range of mutant phenotypes including disruption of the specification of R8 photoreceptor cell diversity. We demonstrate that hibris is required within the retina, and that hibris over-expression is sufficient to disrupt normal photoreceptor cell patterning. These findings suggest an additional layer of complexity in the signaling process that produces paired expression of opsin genes in adjacent R7 and R8 photoreceptor cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Mutation , Organ Specificity , Photoreceptor Cells, Invertebrate/cytology , Retina/metabolism
7.
Exp Eye Res ; 200: 108254, 2020 11.
Article in English | MEDLINE | ID: mdl-32961174

ABSTRACT

Ongoing research using cell transplantation and viral-mediated gene therapy has been making progress to restore vision by retinal repair, but targeted delivery and complete cellular integration remain challenging. An alternative approach is to induce endogenous Müller glia (MG) to regenerate lost neurons and photoreceptors, as occurs spontaneously in teleost fish and amphibians. Extracellular vesicles (EVs) can transfer protein and RNA cargo between cells serving as a novel means of cell-cell communication. We conducted an in vivo screen in zebrafish to identify sources of EVs that could induce MG to dedifferentiate and generate proliferating progenitor cells after intravitreal injection into otherwise undamaged zebrafish eyes. Small EVs (sEVs) from C6 glioma cells were the most consistent at inducing MG-derived proliferating cells. Ascl1a expression increased after intravitreal injection of C6 sEVs and knockdown of ascl1a inhibited the induction of proliferation. Proteomic and RNAseq analyses of EV cargo content were performed to begin to identify key factors that might target EVs to MG and initiate retina regeneration.


Subject(s)
Extracellular Vesicles , Neurogenesis , Photoreceptor Cells, Invertebrate/metabolism , Proteomics/methods , Retina/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Proliferation , Cells, Cultured , Injections , Photoreceptor Cells, Invertebrate/cytology , Retina/cytology , Zebrafish
8.
Evolution ; 74(9): 2105-2120, 2020 09.
Article in English | MEDLINE | ID: mdl-32716056

ABSTRACT

Morphologically diverse eyes have evolved numerous times, yet little is known about how eye gain and loss is related to photic environment. The pteriomorphian bivalves (e.g., oysters, scallops, and ark clams), with a remarkable range of photoreceptor organs and ecologies, are a suitable system to investigate the association between eye evolution and ecological shifts. The present phylogenetic framework was based on amino acid sequences from transcriptome datasets and nucleotide sequences of five additional genes. In total, 197 species comprising 22 families from all five pteriomorphian orders were examined, representing the greatest taxonomic sampling to date. Morphological data were acquired for 162 species and lifestyles were compiled from the literature for all 197 species. Photoreceptor organs occur in 11 families and have arisen exclusively in epifaunal lineages, that is, living above the substrate, at least five times independently. Models for trait evolution consistently recovered higher rates of loss over gain. Transitions to crevice-dwelling habit appear associated with convergent gains of eyespots in epifaunal lineages. Once photoreceptor organs have arisen, multiple losses occurred in lineages that shift to burrowing lifestyles and deep-sea habitats. The observed patterns suggest that eye evolution in pteriomorphians might have evolved in association with light-guided behaviors, such as phototaxis, body posture, and alarm responses.


Subject(s)
Biological Evolution , Bivalvia/cytology , Photoreceptor Cells, Invertebrate/cytology , Animals , Bivalvia/physiology , Photoreceptor Cells, Invertebrate/physiology , Sequence Analysis, Protein , Sequence Analysis, RNA
9.
PLoS Genet ; 16(6): e1008890, 2020 06.
Article in English | MEDLINE | ID: mdl-32579558

ABSTRACT

The Drosophila apical photoreceptor membrane is defined by the presence of two distinct morphological regions, the microvilli-based rhabdomere and the stalk membrane. The subdivision of the apical membrane contributes to the geometrical positioning and the stereotypical morphology of the rhabdomeres in compound eyes with open rhabdoms and neural superposition. Here we describe the characterization of the photoreceptor specific protein PIP82. We found that PIP82's subcellular localization demarcates the rhabdomeric portion of the apical membrane. We further demonstrate that PIP82 is a phosphorylation target of aPKC. PIP82 localization is modulated by phosphorylation, and in vivo, the loss of the aPKC/Crumbs complex results in an expansion of the PIP82 localization domain. The absence of PIP82 in photoreceptors leads to misshapped rhabdomeres as a result of misdirected cellular trafficking of rhabdomere proteins. Comparative analyses reveal that PIP82 originated de novo in the lineage leading to brachyceran Diptera, which is also characterized by the transition from fused to open rhabdoms. Taken together, these findings define a novel factor that delineates and maintains a specific apical membrane domain, and offers new insights into the functional organization and evolutionary history of the Drosophila retina.


Subject(s)
Cell Membrane/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/genetics , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Animals, Genetically Modified , Biological Evolution , Cell Differentiation/genetics , Cell Membrane/genetics , Cell Membrane/ultrastructure , Cell Polarity/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Female , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Function Mutation , Male , Microscopy, Electron, Transmission , Phosphorylation , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/ultrastructure , Phylogeny , Protein Kinase C/metabolism , Retina/cytology , Retina/ultrastructure , Transcription, Genetic
10.
Mol Brain ; 13(1): 1, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31900209

ABSTRACT

The establishment of the functional nervous system requires coordinated development of neurons and glia in the embryo. Our understanding of underlying molecular and cellular mechanisms, however, remains limited. The developing Drosophila visual system is an excellent model for understanding the developmental control of the nervous system. By performing a systematic transgenic RNAi screen, we investigated the requirements of secreted proteins and cell-surface receptors for the development of photoreceptor neurons (R cells) and wrapping glia (WG) in the Drosophila visual system. From the screen, we identified seven genes whose knockdown disrupted the development of R cells and/or WG, including amalgam (ama), domeless (dome), epidermal growth factor receptor (EGFR), kuzbanian (kuz), N-Cadherin (CadN), neuroglian (nrg), and shotgun (shg). Cell-type-specific analysis revealed that ama is required in the developing eye disc for promoting cell proliferation and differentiation, which is essential for the migration of glia in the optic stalk. Our results also suggest that nrg functions in both eye disc and WG for coordinating R-cell and WG development.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Compound Eye, Arthropod/growth & development , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Imaginal Discs/metabolism , Immunoglobulins/physiology , Neurogenesis/genetics , Neuroglia/metabolism , Neurons/metabolism , RNA Interference , Animals , Animals, Genetically Modified , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Cell Lineage , Cell Movement , Compound Eye, Arthropod/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Gene Knockdown Techniques , Imaginal Discs/cytology , Immunoglobulins/genetics , Larva , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism
11.
J Comp Neurol ; 528(10): 1754-1774, 2020 07.
Article in English | MEDLINE | ID: mdl-31860126

ABSTRACT

The compound eye of cockroaches is obligatory for entrainment of the Madeira cockroach's circadian clock, but the cellular nature of its entrainment pathways is enigmatic. Employing multiple-label immunocytochemistry, histochemistry, and backfills, we searched for photic entrainment pathways to the accessory medulla (AME), the circadian clock of the Madeira cockroach. We wanted to know whether photoreceptor terminals could directly contact pigment-dispersing factor-immunoreactive (PDF-ir) circadian pacemaker neurons with somata in the lamina (PDFLAs) or somata next to the AME (PDFMEs). Short green-sensitive photoreceptor neurons of the compound eye terminated in lamina layers LA1 and LA2, adjacent to PDFLAs and PDFMEs that branched in LA3. Long UV-sensitive compound eye photoreceptor neurons terminated in medulla layer ME2 without direct contact to ipsilateral PDFMEs that arborized in ME4. Multiple neuropeptide-ir interneurons branched in ME4, connecting the AME to ME2. Before, extraocular photoreceptors of the lamina organ were suggested to send terminals to accessory laminae. There, they overlapped with PDFLAs that mostly colocalized PDF, FMRFamide, and 5-HT immunoreactivities, and with terminals of ipsi- and contralateral PDFMEs. We hypothesize that during the day cholinergic activation of the largest PDFME via lamina organ photoreceptors maintains PDF release orchestrating phases of sleep-wake cycles. As ipsilateral PDFMEs express excitatory and contralateral PDFMEs inhibitory PDF autoreceptors, diurnal PDF release keeps both PDF-dependent clock circuits in antiphase. Future experiments will test whether ipsilateral PDFMEs are sleep-promoting morning cells, while contralateral PDFMEs are activity-promoting evening cells, maintaining stable antiphase via the largest PDFME entrained by extraocular photoreceptors of the lamina organ.


Subject(s)
Circadian Clocks , Neural Pathways/cytology , Neuropil/cytology , Optic Lobe, Nonmammalian/cytology , Photoreceptor Cells, Invertebrate/cytology , Animals , Cockroaches
12.
Elife ; 82019 11 06.
Article in English | MEDLINE | ID: mdl-31692445

ABSTRACT

Drosophila R7 UV photoreceptors (PRs) are divided into yellow (y) and pale (p) subtypes. yR7 PRs express the Dpr11 cell surface protein and are presynaptic to Dm8 amacrine neurons (yDm8) that express Dpr11's binding partner DIP-γ, while pR7 PRs synapse onto DIP-γ-negative pDm8. Dpr11 and DIP-γ expression patterns define 'yellow' and 'pale' color vision circuits. We examined Dm8 neurons in these circuits by electron microscopic reconstruction and expansion microscopy. DIP-γ and dpr11 mutations affect the morphologies of yDm8 distal ('home column') dendrites. yDm8 neurons are generated in excess during development and compete for presynaptic yR7 PRs, and interactions between Dpr11 and DIP-γ are required for yDm8 survival. These interactions also allow yDm8 neurons to select yR7 PRs as their appropriate home column partners. yDm8 and pDm8 neurons do not normally compete for survival signals or R7 partners, but can be forced to do so by manipulation of R7 subtype fate.


Subject(s)
Amacrine Cells/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Membrane Proteins/genetics , Photoreceptor Cells, Invertebrate/metabolism , Synapses/metabolism , Visual Pathways/physiology , Amacrine Cells/cytology , Animals , Color Vision/physiology , Dendrites/metabolism , Dendrites/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Gene Expression , Membrane Proteins/metabolism , Mutation , Photoreceptor Cells, Invertebrate/cytology , Protein Binding , Synapses/ultrastructure , Visual Pathways/cytology
13.
G3 (Bethesda) ; 9(12): 4007-4017, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31649044

ABSTRACT

Drosophila melanogaster photoreceptor cells are highly polarized epithelial cells. Their apical membrane is further subdivided into the stalk membrane and the light-sensing rhabdomere. The photo-pigment Rhodopsin1 (Rh1) localizes to the rhabdomere, whereas the apical determinant Crumbs (Crb) is enriched at the stalk membrane. The proteoglycan Eyes shut (Eys) is secreted through the apical membrane into an inter-rhabdomeral space. Rh1, Crb, and Eys are essential for the development of photoreceptor cells, normal vision, and photoreceptor cell survival. Human orthologs of all three proteins have been linked to retinal degenerative diseases. Here, we describe an RNAi-based screen examining the importance of 237 trafficking-related genes in apical trafficking of Eys, Rh1, and Crb. We found 28 genes that have an effect on the localization and/or levels of these apical proteins and analyzed several factors in more detail. We show that the Arf GEF protein Sec71 is required for biosynthetic traffic of both apical and basolateral proteins, that the exocyst complex and the microtubule-based motor proteins dynein and kinesin promote the secretion of Eys and Rh1, and that Syntaxin 7/Avalanche controls the endocytosis of Rh1, Eys, and Crb.


Subject(s)
Cell Polarity/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genes, Insect , Photoreceptor Cells, Invertebrate/cytology , Animals , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Drosophila Proteins/metabolism , Drosophila melanogaster/ultrastructure , Endocytosis , Exocytosis , Microtubules/metabolism , Photoreceptor Cells, Invertebrate/ultrastructure , Protein Biosynthesis , Protein Transport , RNA Interference
14.
Science ; 366(6463)2019 10 18.
Article in English | MEDLINE | ID: mdl-31582524

ABSTRACT

Sensory systems use stochastic fate specification to increase their repertoire of neuronal types. How these stochastic decisions are coordinated with the development of their targets is unknown. In the Drosophila retina, two subtypes of ultraviolet-sensitive R7 photoreceptors are stochastically specified. In contrast, their targets in the brain are specified through a deterministic program. We identified subtypes of the main target of R7, the Dm8 neurons, each specific to the different subtypes of R7s. Dm8 subtypes are produced in excess by distinct neuronal progenitors, independently from R7. After matching with their cognate R7, supernumerary Dm8s are eliminated by apoptosis. Two interacting cell adhesion molecules, Dpr11 and DIPγ, are essential for the matching of one of the synaptic pairs. These mechanisms allow the qualitative and quantitative matching of R7 and Dm8 and thereby permit the stochastic choice made in R7 to propagate to the brain.


Subject(s)
Drosophila melanogaster/physiology , Neurons/physiology , Photoreceptor Cells, Invertebrate/physiology , Animals , Apoptosis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Communication , Cell Lineage , Cell Shape , Cell Survival , Color Vision , Compound Eye, Arthropod/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Mutation , Neurons/cytology , Photoreceptor Cells, Invertebrate/cytology , Stochastic Processes , Synapses/physiology
15.
Development ; 146(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31371377

ABSTRACT

Drosophila photoreceptors develop from polarized epithelial cells that have apical and basolateral membranes. During morphogenesis, the apical membranes subdivide into a united bundle of photosensory microvilli (rhabdomeres) and a surrounding supporting membrane (stalk). By EMS-induced mutagenesis screening, we found that the F-Bin/Amphiphysin/Rvs (F-BAR) protein syndapin is essential for apical membrane segregation. The analysis of the super-resolution microscopy, STORM and the electron microscopy suggest that syndapin localizes to the neck of the microvilli at the base of the rhabdomere. Syndapin and moesin are required to constrict the neck of the microvilli to organize the membrane architecture at the base of the rhabdomere, to exclude the stalk membrane. Simultaneous loss of syndapin along with the microvilli adhesion molecule chaoptin significantly enhanced the disruption of stalk-rhabdomere segregation. However, loss of the factors involving endocytosis do not interfere. These results indicated syndapin is most likely functioning through its membrane curvature properties, and not through endocytic processes for stalk-rhabdomere segregation. Elucidation of the mechanism of this unconventional domain formation will provide novel insights into the field of cell biology.


Subject(s)
Carrier Proteins/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Microvilli/physiology , Photoreceptor Cells, Invertebrate/physiology , Animals , Carrier Proteins/genetics , Drosophila/genetics , Drosophila/ultrastructure , Drosophila Proteins/genetics , Female , Male , Membrane Proteins/physiology , Microvilli/ultrastructure , Morphogenesis , Mutation , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/ultrastructure
16.
Gene Expr Patterns ; 33: 20-36, 2019 09.
Article in English | MEDLINE | ID: mdl-31096022

ABSTRACT

We exploited the high Ras activity induced differentiation of supernumerary R7 cells in Drosophila eyes to examine if hsrω lncRNAs influence active Ras signaling. Surprisingly, either down- or up-regulation of hsrω lncRNAs in sev-GAL4>RasV12 expressing eye discs resulted in complete pupal lethality and substantially greater increase in R7 photoreceptor number at the expense of cone cells. Enhanced nuclear p-MAPK and presence of sev-GAL4 driven RasV12 bound RafRBDFLAG in cells not expressing the sev-GAL4 driver indicated non-cell autonomous spread of Ras signaling when hsrω levels were co-altered. RNA-sequencing revealed that down-and up-regulation of hsrω transcripts in sev-GAL4>RasV12 expressing eye discs elevated transcripts of positive or negative modulators, respectively, of Ras signaling so that either condition enhances it. Altered hsrω transcript levels in sev-GAL4>RasV12 expressing discs also affected sn/sno/sca RNAs and some other RNA processing transcript levels. Post-transcriptional changes due to the disrupted intra-cellular dynamicity of omega speckle associated hnRNPs and other RNA-binding proteins that follow down- or up-regulation of hsrω lncRNAs appear to be responsible for the further elevated Ras signaling. Cell autonomous and non-autonomous enhancement of Ras signaling by lncRNAs like hsrω has implications for cell signaling during high Ras activity commonly associated with some cancers.


Subject(s)
Drosophila Proteins/genetics , Photoreceptor Cells, Invertebrate/metabolism , RNA, Long Noncoding/genetics , ras Proteins/genetics , Animals , Cell Differentiation , Drosophila Proteins/metabolism , Drosophila melanogaster , MAP Kinase Signaling System , Photoreceptor Cells, Invertebrate/cytology , RNA, Long Noncoding/metabolism , ras Proteins/metabolism
17.
Vision Res ; 158: 100-108, 2019 05.
Article in English | MEDLINE | ID: mdl-30826353

ABSTRACT

Many insects have triplets of camera type eyes, called ocelli, whose function remains unclear for most species. Here, we investigate the ocelli of the bumblebee, Bombus terrestris, using reconstructed 3D data from X-ray microtomography scans combined with computational ray-tracing simulations. This method enables us, not only to predict the visual fields of the ocelli, but to explore for the first time the effect that hair has on them as well as the difference between worker female and male ocelli. We find that bumblebee ocellar fields of view are directed forward and dorsally, incorporating the horizon as well as the sky. There is substantial binocular overlap between the median and lateral ocelli, but no overlap between the two lateral ocelli. Hairs in both workers and males occlude the ocellar field of view, mostly laterally in the worker median ocellus and dorsally in the lateral ocelli. There is little to no sexual dimorphism in the ocellar visual field, suggesting that in B. terrestris they confer no advantage to mating strategies. We compare our results with published observations for the visual fields of compound eyes in the same species as well as with the ocellar vision of other bee and insect species.


Subject(s)
Bees/physiology , Hair/physiology , Photoreceptor Cells, Invertebrate/cytology , Vision, Ocular/physiology , Visual Fields/physiology , Animals , Bees/ultrastructure , Female , Male , Photoreceptor Cells, Invertebrate/ultrastructure , Sex Factors , X-Ray Microtomography
18.
Neuron ; 101(5): 894-904.e5, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30711355

ABSTRACT

Stereotyped synaptic connections define the neural circuits of the brain. In vertebrates, stimulus-independent activity contributes to neural circuit formation. It is unknown whether this type of activity is a general feature of nervous system development. Here, we report patterned, stimulus-independent neural activity in the Drosophila visual system during synaptogenesis. Using in vivo calcium, voltage, and glutamate imaging, we found that all neurons participate in this spontaneous activity, which is characterized by brain-wide periodic active and silent phases. Glia are active in a complementary pattern. Each of the 15 of over 100 specific neuron types in the fly visual system examined exhibited a unique activity signature. The activity of neurons that are synaptic partners in the adult was highly correlated during development. We propose that this cell-type-specific activity coordinates the development of the functional circuitry of the adult brain.


Subject(s)
Action Potentials , Neurogenesis , Photoreceptor Cells, Invertebrate/cytology , Synapses/physiology , Synaptic Potentials , Animals , Calcium/metabolism , Drosophila melanogaster , Glutamic Acid/metabolism , Neuroglia/cytology , Neuroglia/physiology , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/physiology , Visual Pathways/cytology , Visual Pathways/metabolism , Visual Pathways/physiology
19.
Neuroscience ; 396: 66-72, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30458219

ABSTRACT

Drosophila phototransduction occurs in light-sensitive microvilli arranged in a longitudinal structure of the photoreceptor, termed the rhabdomere. Rhodopsin (Rh), isomerized by light, couples to G-protein, which activates phospholipase C (PLC), which in turn cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) generating diacylglycerol (DAG), inositol trisphosphate and H+. This pathway opens the light-dependent channels, transient receptor potential (TRP) and transient receptor potential like (TRPL). PLC and TRP are held together in a protein assembly by the scaffold protein INAD. We report that the channels can be photoactivated in on-cell rhabdomeric patches and in excised patches by DAG. In excised patches, addition of PLC-activator, m-3M3FBS, or G-protein-activator, GTP-γ-S, opened TRP. These reagents were ineffective in PLC-mutant norpA and in the presence of PLC inhibitor U17322. However, DAG activated TRP even when PLC was pharmacologically or mutationally suppressed. These observations indicate that PLC, G-protein, and TRP were retained functional in these patches. DAG also activated TRP in the protein kinase C (PKC) mutant, inaC, excluding the possibility that PKC could mediate DAG-dependent TRP activation. Labeling diacylglycerol kinase (DGK) by fusion of fluorescent mCherry (mCherry-DGK) indicates that DGK, which returns DAG to dark levels, is highly expressed in the microvilli. In excised patches, TRP channels could be light-activated in the presence of GTP, which is required for G-protein activation. The evidence indicates that the proteins necessary for phototransduction are retained functionally after excision and that DAG is necessary and sufficient for TRP opening. This work opens up unique possibilities for studying, in sub-microscopic native membrane patches, the ubiquitous phosphoinositide signaling pathway and its regulatory mechanisms in unprecedented detail.


Subject(s)
Ion Channel Gating/radiation effects , Light , Microvilli/metabolism , Microvilli/radiation effects , Photoreceptor Cells, Invertebrate/cytology , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/radiation effects , Animals , Diacylglycerol Kinase/biosynthesis , Diglycerides/pharmacology , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Drosophila Proteins/metabolism , Drosophila Proteins/radiation effects , Drosophila melanogaster , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Membrane Potentials/drug effects , Protein Kinase C/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Transient Receptor Potential Channels/isolation & purification , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics
20.
Dev Biol ; 445(2): 245-255, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30502325

ABSTRACT

The ascidian larva has a pigmented ocellus comprised of a cup-shaped array of approximately 30 photoreceptor cells, a pigment cell, and three lens cells. Morphological, physiological and molecular evidence has suggested evolutionary kinship between the ascidian larval photoreceptors and vertebrate retinal and/or pineal photoreceptors. Rx, an essential factor for vertebrate photoreceptor development, has also been suggested to be involved in the development of the ascidian photoreceptor cells, but a recent revision of the photoreceptor cell lineage raised a crucial discrepancy between the reported expression patterns of Rx and the cell lineage. Here, we report spatio-temporal expression patterns of Rx at single-cell resolution along with mitotic patterns up to the final division of the photoreceptor-lineage cells in Ciona. The expression of Rx commences in non-photoreceptor a-lineage cells on the right side of the anterior sensory vesicle at the early tailbud stage. At the mid tailbud stage, Rx begins to be expressed in the A-lineage photoreceptor cell progenitors located on the right side of the posterior sensory vesicle. Thus, Rx is specifically but not exclusively expressed in the photoreceptor-lineage cells in the ascidian embryo. Two cis-regulatory modules are shown to be important for the photoreceptor-lineage expression of Rx. The cell division patterns of the photoreceptor-lineage cells rationally explain the generation of the cup-shaped structure of the pigmented ocellus. The present findings demonstrate the complete cell lineage of the ocellus photoreceptor cells and provide a framework elucidating the molecular and cellular mechanisms of photoreceptor development in Ciona.


Subject(s)
Ciona intestinalis/growth & development , Ciona intestinalis/genetics , Homeodomain Proteins/genetics , Photoreceptor Cells, Invertebrate/cytology , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , Cell Lineage/genetics , Ciona intestinalis/cytology , Evolution, Molecular , Gene Expression Regulation, Developmental , Larva/cytology , Larva/genetics , Larva/growth & development , Mitosis/genetics , Regulatory Sequences, Ribonucleic Acid , Spatio-Temporal Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...