Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
J Neurogenet ; 22(2): 1, 2008.
Article in English | MEDLINE | ID: mdl-18428030

ABSTRACT

We have identified helmsman (hlm), which is expressed in the fruit fly photoreceptor cells during neural network development. Hlm is also expressed in the elongating cells of the embryonic trachea. Both photoreceptor neurons and embryonic trachea cells elongate in precise, targeted growth for cell-to-cell specific recognition. Expression of antisense hlm-interfering RNA during embryogenesis arrests elongation of the developing tracheal cells and blocks maturation. Expression of hlm-interfering RNA during visual system formation results in reduced visual acuity and poor performance in optomotor response, indicative of abnormal neural network development. Hlm is a unique cell surface protein with complement-like protein interaction motifs. We have also cloned hlm from Lucilia cuprina (Australian blowfly), which is approximately 100 million years divergent from Drosophila, and find a remarkable 90% protein identity over the entire 558 amino acid protein. Analysis of the hlm sequence found in other species indicates a significant evolutionary pressure to maintain the hlm protein sequence. Our interpretation is that hlm is involved in cell maturation in both the elongating trachea and elongating photoreceptor cells. Cell adhesion and cell signaling, which are known to use immunoglobulin-like cell adhesion molecules, may use molecular systems analogous to complement to create protein complexes to regulate growth.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Membrane Proteins/metabolism , Photoreceptor Cells/metabolism , Trachea/metabolism , Amidohydrolases , Amino Acid Sequence , Animals , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Embryo, Nonmammalian/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Insect Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Molecular Sequence Data , Motion Perception/physiology , Photoreceptor Cells/embryology , RNA Interference , Sequence Alignment , Sequence Analysis, DNA/methods , Species Specificity , Trachea/embryology , Visual Acuity/physiology
2.
J Neurosci ; 28(13): 3392-403, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18367606

ABSTRACT

The retinal ganglion cells (RGCs) are the sole output neurons in the retina that form the optic nerve and convey light signals detected by photoreceptors to the higher visual system. Their degeneration and damage caused by glaucoma and injury can lead to blindness. During retinogenesis, RGCs are specified from a population of multipotential precursors capable of generating RGC, amacrine, horizontal, and cone cells. How the RGC fate is selected from these multiple neuron fates is unknown at present. Here we show that the previously unsuspected POU domain transcription factor Brn3b (brain-specific homeobox/POU domain protein 3b) plays such a critical role. Loss of Brn3b function in mice leads to misspecification of early RGC precursors as late-born RGC, amacrine, and horizontal cells, whereas misexpressed Brn3b suppresses non-RGC cell fates but promotes the RGC fate. Microarray profiling and other molecular analyses reveal that, in RGC precursors, Brn3b normally represses the expression of a network of retinogenic factor genes involved in fate commitment and differentiation of late-born RGC, amacrine, horizontal, and cone cells. Our data suggest that Brn3b specifies the RGC fate from multipotential precursors not only by promoting RGC differentiation but also by suppressing non-RGC differentiation programs as a safeguard mechanism.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Retina/cytology , Retinal Ganglion Cells/physiology , Transcription Factor Brn-3B/physiology , Amacrine Cells/metabolism , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cluster Analysis , Embryo, Mammalian , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Lac Operon/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , Photoreceptor Cells/embryology , Transcription Factor Brn-3B/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Development ; 135(3): 513-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18171686

ABSTRACT

The Drosophila Activin-like ligands Activin-beta and Dawdle control several aspects of neuronal morphogenesis, including mushroom body remodeling, dorsal neuron morphogenesis and motoneuron axon guidance. Here we show that the same two ligands act redundantly through the Activin receptor Babo and its transcriptional mediator Smad2 (Smox), to regulate neuroblast numbers and proliferation rates in the developing larval brain. Blocking this pathway results in the development of larvae with small brains and aberrant photoreceptor axon targeting, and restoring babo function in neuroblasts rescued these mutant phenotypes. These results suggest that the Activin signaling pathway is required for producing the proper number of neurons to enable normal connection of incoming photoreceptor axons to their targets. Furthermore, as the Activin pathway plays a key role in regulating propagation of mouse and human embryonic stem cells, our observation that it also regulates neuroblast numbers and proliferation in Drosophila suggests that involvement of Activins in controlling stem cell propagation may be a common regulatory feature of this family of TGF-beta-type ligands.


Subject(s)
Activins/metabolism , Brain/cytology , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Activin Receptors/metabolism , Activins/genetics , Animals , Axons/metabolism , Brain/embryology , Carrier Proteins/genetics , Cell Count , Cell Proliferation , Cyclin A/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Larva/cytology , Mitosis , Mutation/genetics , Photoreceptor Cells/cytology , Photoreceptor Cells/embryology , Retina/cytology , Retina/metabolism , S Phase , Signal Transduction , Smad2 Protein/metabolism , Stem Cells/cytology , Superior Colliculi/embryology
4.
J Comp Neurol ; 506(4): 584-603, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18072193

ABSTRACT

In the retina of warm-blooded vertebrates, photoreceptors are specified many days before the onset of synaptogenesis and the expression of photopigments. The factors that regulate the maturation of photoreceptors in the developing retina remain unknown. We report here that photoreceptors transiently express LIM-domain transcription factors during the development of the chicken retina. We examined the differentiation of photoreceptors through the normal course of embryonic development and at the far periphery of the postnatal retina, where the differentiation of photoreceptors is slowed and persists across a spatial gradient. In the embryonic retina, we find visinin-positive photoreceptors that transiently express Islet2 and Lim3 starting at E8 and ending around E15, but persisting in far peripheral regions of the retina through the first 2 weeks of postnatal development. During early stages of photoreceptor maturation, there is coincident and transient expression of the LIM-domain factors with axonin1, a cell surface glycoprotein that is a member of the immunoglobulin superfamily. Coincident with the downregulation of Islet2 and Lim3, we find the upregulation of calbindin, red/green opsin, rhodopsin, and a synaptic marker in the outer plexiform layer (OPL; dystrophin). In the periphery of the postnatal retina, photoreceptors that express Islet2, Lim3, and axonin1 do not overlap with photoreceptors that express calbindin, red/green opsin, rhodopsin, and dystrophin. We propose that Islet2 and Lim3 may promote the expression of genes that are involved in the early stages of differentiation but may suppress the expression of genes that are required in the mature photoreceptors.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Photoreceptor Cells/embryology , Photoreceptor Cells/growth & development , Transcription Factors/metabolism , Aging/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Chick Embryo , Chickens , Contactin 2 , Down-Regulation/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells/metabolism , Rod Opsins/genetics , Rod Opsins/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
J Comp Neurol ; 506(5): 822-37, 2008 Feb 10.
Article in English | MEDLINE | ID: mdl-18076030

ABSTRACT

The cellular and molecular mechanisms underlying photoreceptor synaptogenesis are poorly understood. Furthermore, a detailed picture of the molecular composition of photoreceptor synapses, or their subtypes, is not yet available, nor do we know what differences, if any, exist among those subtypes. To address these questions, we investigated temporal and spatial patterns of expression and assembly of photoreceptor presynaptic components during chick embryo retinal development and early posthatched life by using reverse transcriptase polymerase chain reaction (RT-PCR), dissociated retinal cells, laser-capture microdissection (LCM), immunocytochemistry and confocal microscopy. Immunocytochemistry in tissue sections and dissociated cells showed many similarities and few differences in the synaptic composition of rods and cone subtypes, which, however, were found to project to different strata within the outer plexiform layer. A striking finding was the precise timetable of expression of synaptic genes and proteins during synaptogenesis. Although mRNAs for some synaptic molecules appeared as early as embryonic day (ED) 5-8 (the time of inner retina synaptogenesis), others were undetectable before the time of onset of photoreceptor synaptogenesis on ED13, including CAST, rim2, synapsin-2, syntaxin-3, synaptotagmin, glutamate receptors -1, -4, and -5, homer-1 and -2, and tenascin-R. Most synaptic proteins in photoreceptors followed a similar sequence of expression: they were negative or weakly positive before ED13, appeared in inner segments between ED13 and ED15, became subsequently detectable in perinuclear and axonal regions, and by ED18 were assembled into synaptic terminals and became undetectable in the inner segments. The identity of the signals that regulate the coordinated expression of these synaptic components remains to be investigated.


Subject(s)
Gene Expression Profiling , Photoreceptor Cells/embryology , Retina/embryology , Synapses/metabolism , Tissue Distribution/physiology , Animals , Chick Embryo , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Nerve Tissue Proteins/metabolism , Organogenesis , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Presynaptic Terminals/classification , Presynaptic Terminals/metabolism , Retina/cytology , Retina/metabolism , Synapses/classification , Time Factors
6.
Dev Dyn ; 237(1): 286-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18095352

ABSTRACT

Muscleblind-like (MBNL) is a CCCH zinc finger-containing RNA-binding protein required for the development of both muscle and photoreceptors in Drosophila; it is conserved evolutionarily, and it is associated in humans with the muscular disease myotonic dystrophy. Its role in the development of vertebrate retinal cells, however, remains unknown. As an initial approach to its investigation, we have cloned three chick muscleblind genes, characterized their isoforms, and examined their expression patterns in the chick embryo retina. The relative levels of expression of the MBNL genes increased during embryonic development. In situ hybridization (ISH) showed that the three MBNL mRNAs had widespread patterns of expression at all the developmental stages examined. Of interest, the temporal and spatial patterns of protein expression, detected by immunocytochemistry with antibodies against MBNL1 and MBNL2, were much more restricted than those seen by ISH. At early stages (ED5-7), for example, MBNL1 and MBNL2 mRNAs were present throughout the retina, but immunoreactivity for the corresponding proteins was largely restricted to the periphery of the optic cup (presumptive iris/ciliary epithelium/ciliary margin zone). MBNL1 and MBNL2 immunoreactivity became detectable at the fundus at later stages, but was limited to a very small subset of the cells that had ISH signals for the cognate mRNAs (particularly ganglion cells and photoreceptors). Within photoreceptors, MBNL1 and MBNL2 immunoreactivity first appeared in their inner segments; MBNL2 remained there, but MBNL1 became subsequently localized to their synaptic terminals. These expression patterns are consistent with the possibility that MBNLs may regulate photoreceptor development in the chick retina, much as MBL does in Drosophila, and suggest that the expression of MBNL1 and MBNL2 may be regulated posttranscriptionally.


Subject(s)
Gene Expression Regulation, Developmental , RNA-Binding Proteins/genetics , Retina/metabolism , Alternative Splicing , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation/genetics , Chick Embryo , Chickens , In Situ Hybridization , Microscopy, Confocal , Microscopy, Fluorescence , Photoreceptor Cells/cytology , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , RNA-Binding Proteins/metabolism , Retina/embryology
7.
Brain Res ; 1192: 114-33, 2008 Feb 04.
Article in English | MEDLINE | ID: mdl-17662965

ABSTRACT

Rod and cone photoreceptors in the mammalian retina are special types of neurons that are responsible for phototransduction, the first step of vision. Development and maintenance of photoreceptors require precisely regulated gene expression. This regulation is mediated by a network of photoreceptor transcription factors centered on Crx, an Otx-like homeodomain transcription factor. The cell type (subtype) specificity of this network is governed by factors that are preferentially expressed by rods or cones or both, including the rod-determining factors neural retina leucine zipper protein (Nrl) and the orphan nuclear receptor Nr2e3; and cone-determining factors, mostly nuclear receptor family members. The best-documented of these include thyroid hormone receptor beta2 (Tr beta2), retinoid related orphan receptor Ror beta, and retinoid X receptor Rxr gamma. The appropriate function of this network also depends on general transcription factors and cofactors that are ubiquitously expressed, such as the Sp zinc finger transcription factors and STAGA co-activator complexes. These cell type-specific and general transcription regulators form complex interactomes; mutations that interfere with any of the interactions can cause photoreceptor development defects or degeneration. In this manuscript, we review recent progress on the roles of various photoreceptor transcription factors and interactions in photoreceptor subtype development. We also provide evidence of auto-, para-, and feedback regulation among these factors at the transcriptional level. These protein-protein and protein-promoter interactions provide precision and specificity in controlling photoreceptor subtype-specific gene expression, development, and survival. Understanding these interactions may provide insights to more effective therapeutic interventions for photoreceptor diseases.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Feedback, Physiological/genetics , Feedback, Physiological/physiology , Humans , Photoreceptor Cells/cytology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/physiopathology , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism
8.
Brain Res ; 1192: 5-16, 2008 Feb 04.
Article in English | MEDLINE | ID: mdl-17692298

ABSTRACT

Evolutionary and other functional accounts of the retina and its normal development highlight different aspects of control of its growth and form than genomic and mechanistic accounts. Discussing examples from opsin expression, developmental regulation of the eye's size and optical quality, regulation of eye size with respect to brain and body size, and the development of the fovea, these different aspects of control are contrasted. Contributions of mouse models, particularly with regard to relative timing of events in different species are reviewed, introducing a Web-based utility for exploration of timing issues (www.translatingtime.net). Variation at the individual level, in early experience, and also across species is an essential source of information to understand normal development and its pathologies.


Subject(s)
Cell Differentiation/genetics , Retina/embryology , Retina/growth & development , Rod Opsins/genetics , Animals , Biological Evolution , Dark Adaptation/genetics , Fovea Centralis/cytology , Fovea Centralis/embryology , Fovea Centralis/growth & development , Humans , Mice , Models, Animal , Photoreceptor Cells/cytology , Photoreceptor Cells/embryology , Photoreceptor Cells/growth & development , Retina/cytology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism
9.
Brain Res ; 1192: 134-50, 2008 Feb 04.
Article in English | MEDLINE | ID: mdl-17466954

ABSTRACT

How does a retinal progenitor choose to differentiate as a rod or a cone and, if it becomes a cone, which one of their different subtypes? The mechanisms of photoreceptor cell fate specification and differentiation have been extensively investigated in a variety of animal model systems, including human and non-human primates, rodents (mice and rats), chickens, frogs (Xenopus) and fish. It appears timely to discuss whether it is possible to synthesize the resulting information into a unified model applicable to all vertebrates. In this review we focus on several widely used experimental animal model systems to highlight differences in photoreceptor properties among species, the diversity of developmental strategies and solutions that vertebrates use to create retinas with photoreceptors that are adapted to the visual needs of their species, and the limitations of the methods currently available for the investigation of photoreceptor cell fate specification. Based on these considerations, we conclude that we are not yet ready to construct a unified model of photoreceptor cell fate specification in the developing vertebrate retina.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Regulation, Enzymologic/genetics , Photoreceptor Cells/embryology , Stem Cells/metabolism , Vertebrates/embryology , Animals , Biological Evolution , Humans , Models, Animal , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Phylogeny , Species Specificity , Stem Cells/cytology
10.
Brain Res ; 1192: 151-64, 2008 Feb 04.
Article in English | MEDLINE | ID: mdl-17553468

ABSTRACT

Although the neural retina appears as a relatively uniform tissue when viewed from its surface, it is in fact highly patterned along its anterior-posterior and dorso-ventral axes. The question of how and when such patterns arise has been the subject of intensive investigations over several decades. Most studies aimed at understanding retinal pattern formation have used the retinotectal map, the ordered projections of retinal ganglion cells to the brain, as a functional readout of the pattern. However, other cell types are also topographically organized in the retina. The most commonly recognized example of such a topographic cellular organization is the differential distribution of photoreceptor types across the retina. Photoreceptor patterns are highly species-specific and may represent an important adaptation to the visual niche a given species occupies. Nevertheless, few studies have addressed this functional readout of pattern to date and our understanding of its development has remained superficial. Here, we review recent advances in understanding the molecular cascades that control regionalization of the eye anlage, relate these findings to the development of photoreceptor patterns and discuss common and unique strategies involved in both aspects of retinal pattern formation.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental/genetics , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , Retinal Ganglion Cells/metabolism , Vertebrates/embryology , Animals , Cell Differentiation/genetics , Humans , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Photoreceptor Cells/cytology , Retinal Ganglion Cells/cytology , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism
11.
Indian J Exp Biol ; 45(5): 469-74, 2007 May.
Article in English | MEDLINE | ID: mdl-17569291

ABSTRACT

Cadmium (Cd) is one of the environmental contaminant and because of its non-decomposable character, it can damage nature. In this study, TEM was used in order to assess the ultrastructural effects of Cd on photorececptor and ganglionic cells of mouse retinal layer. Apoptotic nuclei, heterochromatic nuclei, deletion of nucleus membrane, invisible nucleolus, and apoptotic cells with mitochondrial changes were observed in mice embryo (days 15 of gestation) following CdCl2 injection to mothers on day 9 of gestation. Cadmium exposure caused apoptotic changes both in photoreceptors and ganglionic cells.


Subject(s)
Cadmium/toxicity , Ganglia, Sensory/drug effects , Ganglia, Sensory/embryology , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , Retina/drug effects , Retina/embryology , Animals , Apoptosis/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Female , Ganglia, Sensory/ultrastructure , Male , Mice , Mice, Inbred BALB C , Photoreceptor Cells/ultrastructure , Retina/ultrastructure
12.
Pflugers Arch ; 454(5): 805-19, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17226052

ABSTRACT

Phototransduction is the process by which light triggers an electrical signal in a photoreceptor cell. Image-forming vision in vertebrates is mediated by two types of photoreceptors: the rods and the cones. In this review, we provide a summary of the success in which the mouse has served as a vertebrate model for studying rod phototransduction, with respect to both the activation and termination steps. Cones are still not as well-understood as rods partly because it is difficult to work with mouse cones due to their scarcity and fragility. The situation may change, however.


Subject(s)
Photoreceptor Cells/physiology , Vision, Ocular/physiology , Adaptation, Ocular/physiology , Animals , Differential Threshold , Light , Mice , Mice, Mutant Strains , Photoreceptor Cells/cytology , Photoreceptor Cells/embryology , Refractory Period, Electrophysiological , Retina/cytology , Retina/embryology , Retina/physiology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/embryology , Retinal Pigments/metabolism , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/embryology , Rhodopsin/metabolism , Second Messenger Systems/physiology , Transducin/metabolism
13.
EMBO J ; 26(3): 764-74, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17255935

ABSTRACT

Photoreceptor cell-specific nuclear receptor (PNR) (NR2E3) acts as a sequence-specific repressor that controls neuronal differentiation in the developing retina. We identified a novel PNR co-repressor, Ret-CoR, that is expressed in the developing retina and brain. Biochemical purification of Ret-CoR identified a multiprotein complex that included E2F/Myb-associated proteins, histone deacetylases (HDACs) and NCoR/HDAC complex-related components. Ret-CoR appeared to function as a platform protein for the complex, and interacted with PNR via two CoRNR motifs. Purified Ret-CoR complex exhibited HDAC activity, co-repressed PNR transrepression function in vitro, and co-repressed PNR function in PNR target gene promoters, presumably in the retinal progenitor cells. Notably, the appearance of Ret-CoR protein was cell-cycle-stage-dependent (from G1 to S). Therefore, Ret-CoR appears to act as a component of an HDAC co-repressor complex that supports PNR repression function in the developing retina, and may represent a co-regulator class that supports transcriptional regulator function via cell-cycle-dependent expression.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Developmental , Multiprotein Complexes/metabolism , Photoreceptor Cells/embryology , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Histone Deacetylases/metabolism , Mice , Multiprotein Complexes/genetics , Orphan Nuclear Receptors , RNA Helicases , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Repressor Proteins/isolation & purification
14.
Dev Dyn ; 236(1): 142-55, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17075872

ABSTRACT

The contribution of timing cues from the environment to the coordination of early developmental processes is poorly understood. The day-night cycle represents one of the most important, regular environmental changes that animals are exposed to. A key adaptation that allows animals to anticipate daily environmental changes is the circadian clock. In this review, we aim to address when a light-regulated circadian clock first emerges during development and what its functions are at this early stage. In particular, do circadian clocks regulate early developmental processes? We will focus on results obtained with Drosophila and vertebrates, where both circadian clock and developmental control mechanisms have been intensively studied.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Growth and Development , Animals , Biological Clocks/genetics , Cell Proliferation , Circadian Rhythm/genetics , Drosophila/embryology , Drosophila/genetics , Drosophila/physiology , Models, Biological , Photoperiod , Photoreceptor Cells/embryology , Photoreceptor Cells/physiology , Photoreceptor Cells, Vertebrate/physiology
15.
Curr Opin Cell Biol ; 19(1): 51-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17175152

ABSTRACT

In epithelial cells, the Ezrin, Radixin and Moesin (ERM) proteins are involved in many cellular functions, including regulation of actin cytoskeleton, control of cell shape, adhesion and motility, and modulation of signaling pathways. However, discerning the specific cellular roles of ERMs has been complicated by redundancy between these proteins. Recent genetic studies in model organisms have identified unique roles for ERM proteins. These include the regulation of morphogenesis and maintenance of integrity of epithelial cells, stabilization of intercellular junctions, and regulation of the Rho small GTPase. These studies also suggest that ERMs have roles in actomyosin contractility and vesicular trafficking in the apical domain of epithelial cells. Thus, genetic analysis has enhanced our understanding of these widely expressed membrane-associated proteins.


Subject(s)
Cytoskeletal Proteins/physiology , Epithelial Cells/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , Actomyosin/metabolism , Animals , Biological Transport , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Cytoskeletal Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Drosophila/physiology , Enzyme Activation , Intercellular Junctions/physiology , Intestinal Mucosa/embryology , Intestinal Mucosa/physiology , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Photoreceptor Cells/embryology , Photoreceptor Cells/physiology , Secretory Vesicles/physiology , Signal Transduction , rho GTP-Binding Proteins/metabolism
16.
Invest Ophthalmol Vis Sci ; 47(11): 5066-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065528

ABSTRACT

PURPOSE: To study the possibility of generating photoreceptors through programming RPE transdifferentiation by examining cell differentiation after transplantation into the developing chick eye. METHODS: RPE was isolated, and the cells were dissociated, cultured, and guided to transdifferentiate by infection with retrovirus expressing neuroD (RCAS-neuroD), using RCAS-green fluorescence protein (GFP) as a control. The cells were then harvested and microinjected into the developing eyes of day 5 to day 7 chick embryos, and their development and integration were analyzed. RESULTS: Cells from the control culture integrated into the host RPE. When grafted cells were present in large number, multilayered RPE-like tissues were formed, and the extra tissues consisted of grafted cells and host cells. None of the cells from the control culture expressed photoreceptor-specific genes. In contrast, most cells from RCAS-neuroD-infected culture remained depigmented. A large number of them expressed photoreceptor-specific genes, such as visinin and opsins. Antibodies against red opsin decorated the apical tips and the cell bodies of the grafted, transdifferentiating cells. In the subretinal space, visinin(+) cells aligned along the RPE or an RPE-like structure. When integrated into the host outer nuclear layer, grafted cells emanated elaborate, axonal arborization into the outer plexiform layer of the host retina. CONCLUSIONS: Cultured RPE cells retained their remarkable regenerative capabilities. Cells guided to transdifferentiate along the photoreceptor pathway by neuroD developed a highly ordered cellular structure and could integrate into the outer nuclear layer. These data suggest that, through genetic programming, RPE cells could be a potential source of photoreceptor cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/transplantation , Photoreceptor Cells/embryology , Pigment Epithelium of Eye/embryology , Retina/surgery , Animals , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/physiology , Chick Embryo , Embryonic Stem Cells/cytology , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , In Situ Hybridization , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Retina/cytology , Rod Opsins/metabolism
17.
Dev Dyn ; 235(10): 2641-55, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16788994

ABSTRACT

Despite ongoing interest into the architecture, biochemistry, and physiology of the visual systems of the xiphosuran Limulus polyphemus, their ontogenetic aspects have received little attention. Thus, we explored the development of the lateral eyes and associated neuropils in late embryos and larvae of these animals. The first external evidence of the lateral eyes was the appearance of white pigment spots-guanophores associated with the rudimentary photoreceptors-on the dorsolateral side of the late embryos, suggesting that these embryos can perceive light. The first brown pigment emerges in the eyes during the last (third) embryonic molt to the trilobite stage. However, ommatidia develop from this field of pigment toward the end of the larval trilobite stage so that the young larvae at hatching do not have object recognition. Double staining with the proliferation marker bromodeoxyuridine (BrdU) and an antibody against L. polyphemus myosin III, which is concentrated in photoreceptors of this species, confirmed previous reports that, in the trilobite larvae, new cellular material is added to the eye field from an anteriorly located proliferation zone. Pulse-chase experiments indicated that these new cells differentiate into new ommatidia. Examining larval eyes labeled for opsin showed that the new ommatidia become organized into irregular rows that give the eye field a triangular appearance. Within the eye field, the ommatidia are arranged in an imperfect hexagonal array. Myosin III immunoreactivity in trilobite larvae also revealed the architecture of the central visual pathways associated with the median eye complex and the lateral eyes. Double labeling with myosin III and BrdU showed that neurogenesis persists in the larval brain and suggested that new neurons of both the lamina and the medulla originate from a single common proliferation zone. These data are compared with eye development in Drosophila melanogaster and are discussed with regard to new ideas on eye evolution in the Euarthropoda.


Subject(s)
Arthropods/embryology , Biological Evolution , Eye/embryology , Horseshoe Crabs/embryology , Animals , Arrestin/analysis , Arthropods/anatomy & histology , Arthropods/metabolism , Eye/anatomy & histology , Eye/metabolism , Female , Horseshoe Crabs/anatomy & histology , Horseshoe Crabs/metabolism , Immunohistochemistry/methods , Male , Microscopy, Confocal , Microscopy, Fluorescence , Myosin Type III/analysis , Neurons/cytology , Neurons/metabolism , Neuropil/cytology , Neuropil/metabolism , Optic Nerve/cytology , Optic Nerve/embryology , Optic Nerve/metabolism , Photoreceptor Cells/anatomy & histology , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , Visual Pathways/anatomy & histology , Visual Pathways/embryology , Visual Pathways/metabolism
19.
Dev Biol ; 290(2): 411-20, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16406325

ABSTRACT

Dystroglycan (DG) is a transmembrane receptor linking the extracellular matrix to the internal cytoskeleton. Its structural function has been mainly characterized in muscle fibers, but DG plays signaling and developmental roles also in different tissues and cell types. We have investigated the effects of dystroglycan depletion during eye development of Xenopus laevis. We have injected a specific morpholino (Mo) antisense oligonucleotide in the animal pole of one dorsal blastomere of embryos at four cells stage. Mo-mediated loss of DG function caused disruption of the basal lamina layers, increased apoptosis and reduction of the expression domains of specific retinal markers, at early stages. Later in development, morphants displayed unilateral ocular malformations, such as microphtalmia and retinal delayering with photoreceptors and ganglion cells scattered throughout the retina or aggregated in rosette-like structures. These results recall the phenotypes observed in specific human diseases and suggest that DG presence is crucial at early stages for the organization of retinal architecture.


Subject(s)
Dystroglycans/physiology , Gene Expression Regulation, Developmental , Retina/embryology , Animals , Body Patterning , Bromodeoxyuridine/pharmacology , Cytoskeleton/metabolism , Dystroglycans/biosynthesis , Dystroglycans/metabolism , Female , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Microphthalmia-Associated Transcription Factor/metabolism , Oligonucleotides, Antisense/pharmacology , Phenotype , Photoreceptor Cells/embryology , Retinal Ganglion Cells/metabolism , Tissue Distribution , Xenopus , Xenopus laevis
20.
Development ; 132(3): 615-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15634702

ABSTRACT

In the sensory receptors of both the eye and the ear, specialized apical structures have evolved to detect environmental stimuli such as light and sound. Despite the morphological divergence of these specialized structures and differing transduction mechanisms, the receptors appear to rely in part on a shared group of genes for function. For example, mutations in Usher (USH) genes cause a syndrome of visual and acoustic-vestibular deficits in humans. Several of the affected genes have been identified, including the USH1F gene, which encodes protocadherin 15 (PCDH15). Pcdh15 mutant mice also have both auditory and vestibular defects, although visual defects are not evident. Here we show that zebrafish have two closely related pcdh15 genes that are required for receptor-cell function and morphology in the eye or ear. Mutations in pcdh15a cause deafness and vestibular dysfunction, presumably because hair bundles of inner-ear receptors are splayed. Vision, however, is not affected in pcdh15a mutants. By contrast, reduction of pcdh15b activity using antisense morpholino oligonucleotides causes a visual defect. Optokinetic and electroretinogram responses are reduced in pcdh15b morpholino-injected larvae. In electron micrographs, morphant photoreceptor outer segments are improperly arranged, positioned perpendicular to the retinal pigment epithelium and are clumped together. Our results suggest that both cadherins act within their respective transduction organelles: Pcdh15a is necessary for integrity of the stereociliary bundle, whereas Pcdh15b is required for alignment and interdigitation of photoreceptor outer segments with the pigment epithelium. We conclude that after a duplication of pcdh15, one gene retained an essential function in the ear and the other in the eye.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Fish Proteins/metabolism , Genes, Duplicate/genetics , Hearing/physiology , Vision, Ocular/physiology , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Cadherin Related Proteins , Electrophysiology , Fish Proteins/genetics , Gene Duplication , Gene Expression Regulation, Developmental , Hearing/genetics , Microscopy, Electron , Mutation/genetics , Phenotype , Photoreceptor Cells/cytology , Photoreceptor Cells/embryology , Photoreceptor Cells/metabolism , Photoreceptor Cells/ultrastructure , Retina/embryology , Retina/metabolism , Retina/ultrastructure , Sensitivity and Specificity , Vision, Ocular/genetics , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...