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










Publication year range
1.
Invest Ophthalmol Vis Sci ; 64(5): 1, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37126314

ABSTRACT

Purpose: Familial exudative vitreoretinopathy (FEVR) and Norrie disease are examples of genetic disorders in which the retinal vasculature fails to fully form (hypovascular), leading to congenital blindness. While studying the role of a factor expressed during retinal development, T-box factor Tbx3, we discovered that optic cup loss of Tbx3 caused the retina to become hypovascular. The purpose of this study was to characterize how loss of Tbx3 affects retinal vasculature formation. Methods: Conditional removal of Tbx3 from both retinal progenitors and astrocytes was done using the optic cup-Cre recombinase driver BAC-Dkk3-Cre and was analyzed using standard immunohistochemical techniques. Results: With Tbx3 loss, the retinas were hypovascular, as seen in patients with retinopathy of prematurity (ROP) and FEVR. Retinal vasculature failed to form the stereotypic tri-layered plexus in the dorsal-temporal region. Astrocyte precursors were reduced in number and failed to form a lattice at the dorsal-temporal edge. We next examined retinal ganglion cells, as they have been shown to play a critical role in retinal angiogenesis. We found that melanopsin expression and Islet1/2-positive retinal ganglion cells were reduced in the dorsal half of the retina. In previous studies, the loss of melanopsin has been linked to hyaloid vessel persistence, which we also observed in the Tbx3 conditional knockout (cKO) retinas, as well as in infants with ROP or FEVR. Conclusions: To the best of our knowledge, these studies are the first demonstration that Tbx3 is required for normal mammalian eye formation. Together, the results provide a potential genetic model for retinal hypovascular diseases.


Subject(s)
Retinal Degeneration , Retinopathy of Prematurity , Mice , Animals , Infant, Newborn , Humans , Retina , Retinal Ganglion Cells , Retinal Vessels , Familial Exudative Vitreoretinopathies , Mammals , T-Box Domain Proteins
2.
Front Cell Dev Biol ; 10: 1016182, 2022.
Article in English | MEDLINE | ID: mdl-36393832

ABSTRACT

Microphthalmia, anophthalmia, and coloboma (MAC) are congenital ocular malformations causing 25% of childhood blindness. The X-linked disorder Focal Dermal Hypoplasia (FDH) is frequently associated with MAC and results from mutations in Porcn, a membrane bound O-acyl transferase required for palmitoylation of Wnts to activate multiple Wnt-dependent pathways. Wnt/ß-catenin signaling is suppressed in the anterior neural plate for initiation of eye formation and is subsequently required during differentiation of the retinal pigment epithelium (RPE). Non-canonical Wnts are critical for early eye formation in frog and zebrafish. However, it is unclear whether this also applies to mammals. We performed ubiquitous conditional inactivation of Porcn in mouse around the eye field stage. In Porcn CKO , optic vesicles (OV) arrest in growth and fail to form an optic cup. Ventral proliferation is significantly decreased in the mutant OV, with a concomitant increase in apoptotic cell death. While pan-ocular transcription factors such as PAX6, SIX3, LHX2, and PAX2 are present, indicative of maintenance of OV identity, regional expression of VSX2, MITF, OTX2, and NR2F2 is downregulated. Failure of RPE differentiation in Porcn CKO is consistent with downregulation of the Wnt/ß-catenin effector LEF1, starting around 2.5 days after inactivation. This suggests that Porcn inactivation affects signaling later than a potential requirement for Wnts to promote eye field formation. Altogether, our data shows a novel requirement for Porcn in regulating growth and morphogenesis of the OV, likely by controlling proliferation and survival. In FDH patients with ocular manifestations, growth deficiency during early ocular morphogenesis may be the underlying cause for microphthalmia.

3.
Hum Mol Genet ; 29(20): 3373-3387, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33075808

ABSTRACT

Uveal coloboma represents one of the most common congenital ocular malformations accounting for up to 10% of childhood blindness (~1 in 5000 live birth). Coloboma originates from defective fusion of the optic fissure (OF), a transient gap that forms during eye morphogenesis by asymmetric, ventral invagination. Genetic heterogeneity combined with the activity of developmentally regulated genes suggests multiple mechanisms regulating OF closure. The tumor suppressor and FERM domain protein Neurofibromin 2 (NF2) controls diverse processes in cancer, development and regeneration, via Hippo pathway and cytoskeleton regulation. In humans, NF2 mutations can cause ocular abnormalities, including coloboma, however, its actual role in OF closure is unknown. Using conditional inactivation in the embryonic mouse eye, our data indicate that loss of Nf2 function results in a novel underlying cause for coloboma. In particular, mutant eyes show substantially increased retinal pigmented epithelium (RPE) proliferation in the fissure region with concomitant acquisition of RPE cell fate. Cells lining the OF margin can maintain RPE fate ectopically and fail to transition from neuroepithelial to cuboidal shape. In the dorsal RPE of the optic cup, Nf2 inactivation leads to a robust increase in cell number, with local disorganization of the cytoskeleton components F-actin and pMLC2. We propose that RPE hyperproliferation is the primary cause for the observed defects causing insufficient alignment of the OF margins in Nf2 mutants and failure to fuse properly, resulting in persistent coloboma. Our findings indicate that limiting proliferation particularly in the RPE layer is a critical mechanism during OF closure.


Subject(s)
Cell Proliferation , Coloboma/pathology , Eye/pathology , Gene Expression Regulation, Developmental , Neurofibromin 2/physiology , Organogenesis , Retinal Pigment Epithelium/pathology , Animals , Coloboma/etiology , Coloboma/metabolism , Eye/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Pigment Epithelium/metabolism
4.
Dev Cell ; 46(3): 376-387.e7, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30086304

ABSTRACT

During development, neurons undergo apoptosis if they do not receive adequate trophic support from tissues they innervate or when detrimental factors activate the p75 neurotrophin receptor (p75NTR) at their axon ends. Trophic factor deprivation (TFD) or activation of p75NTR in distal axons results in a retrograde degenerative signal. However, the nature of this signal and the regulation of its transport are poorly understood. Here, we identify p75NTR intracellular domain (ICD) and histone deacetylase 1 (HDAC1) as part of a retrograde pro-apoptotic signal generated in response to TFD or ligand binding to p75NTR in sympathetic neurons. We report an unconventional function of HDAC1 in retrograde transport of a degenerative signal and its constitutive presence in sympathetic axons. HDAC1 deacetylates dynactin subunit p150Glued, which enhances its interaction with dynein. These findings define p75NTR ICD as a retrograde degenerative signal and reveal p150Glued deacetylation as a unique mechanism regulating axonal transport.


Subject(s)
Axonal Transport/physiology , Axons/metabolism , Dynactin Complex/metabolism , Histone Deacetylase 1/metabolism , Animals , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism
5.
Cell Rep ; 19(6): 1257-1267, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494873

ABSTRACT

EGF receptor (EGFR) is a critical signaling node throughout life. However, it has not been possible to directly visualize endogenous Egfr in mice. Using CRISPR/Cas9 genome editing, we appended a fluorescent reporter to the C terminus of the Egfr. Homozygous reporter mice appear normal and EGFR signaling is intact in vitro and in vivo. We detect distinct patterns of Egfr expression in progenitor and differentiated compartments in embryonic and adult mice. Systemic delivery of EGF or amphiregulin results in markedly different patterns of Egfr internalization and trafficking in hepatocytes. In the normal intestine, Egfr localizes to the crypt rather than villus compartment, expression is higher in adjacent epithelium than in intestinal tumors, and following colonic injury expression appears in distinct cell populations in the stroma. This reporter, under control of its endogenous regulatory elements, enables in vivo monitoring of the dynamics of Egfr localization and trafficking in normal and disease states.


Subject(s)
ErbB Receptors/genetics , Genes, Reporter , Transgenes , Adult Stem Cells/metabolism , Amphiregulin/metabolism , Animals , Embryo, Mammalian/metabolism , ErbB Receptors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/metabolism , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Mice , Microscopy, Fluorescence/methods , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Invest Ophthalmol Vis Sci ; 57(13): 5253-5262, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27701636

ABSTRACT

PURPOSE: The scaffold protein Axin2 is an antagonist and universal target of the Wnt/ß-catenin pathway. Disruption of Axin2 may lead to developmental eye defects; however, this has not been examined. The purpose of this study was to investigate the role of Axin2 during ocular and extraocular development in mouse. METHODS: Animals heterozygous and homozygous for a Axin2lacZ knock-in allele were analyzed at different developmental stages for reporter expression, morphology as well as for the presence of ocular and extraocular markers using histologic and immunohistochemical techniques. RESULTS: During early eye development, the Axin2lacZ reporter was expressed in the periocular mesenchyme, RPE, and optic stalk. In the developing retina, Axin2lacZ reporter expression was initiated in ganglion cells at late embryonic stages and robustly expressed in subpopulations of amacrine and horizontal cells postnatally. Activation of the Axin2lacZ reporter overlapped with labeling of POU4F1, PAX6, and Calbindin. Germline deletion of Axin2 led to variable ocular phenotypes ranging from normal to severely defective eyes exhibiting microphthalmia, coloboma, lens defects, and expanded ciliary margin. These defects were correlated with abnormal tissue patterning in individual affected tissues, such as the optic fissure margins in the ventral optic cup and in the expanded ciliary margin. CONCLUSIONS: Our results reveal a critical role for Axin2 during ocular development, likely by restricting the activity of the Wnt/ß-catenin pathway.


Subject(s)
Axin Protein/genetics , Eye Diseases/genetics , Eye/growth & development , Gene Expression Regulation, Developmental , Organogenesis/genetics , Alleles , Animals , Axin Protein/biosynthesis , Disease Models, Animal , Eye/metabolism , Eye Diseases/metabolism , Eye Diseases/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Wnt Signaling Pathway
7.
Am J Pathol ; 185(1): 197-213, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25451153

ABSTRACT

Wnt glycoproteins control key processes during development and disease by activating various downstream pathways. Wnt secretion requires post-translational modification mediated by the O-acyltransferase encoded by the Drosophila porcupine homolog gene (PORCN). In humans, PORCN mutations cause focal dermal hypoplasia (FDH, or Goltz syndrome), an X-linked dominant multisystem birth defect that is frequently accompanied by ocular abnormalities such as coloboma, microphthalmia, or even anophthalmia. Although genetic ablation of Porcn in mouse has provided insight into the etiology of defects caused by ectomesodermal dysplasia in FDH, the requirement for Porcn and the actual Wnt ligands during eye development have been unknown. In this study, Porcn hemizygosity occasionally caused ocular defects reminiscent of FDH. Conditional inactivation of Porcn in periocular mesenchyme led to defects in mid- and hindbrain and in craniofacial development, but was insufficient to cause ocular abnormalities. However, a combination of conditional Porcn depletion in optic vesicle neuroectoderm, lens, and neural crest-derived periocular mesenchyme induced severe eye abnormalities with high penetrance. In particular, we observed coloboma, transdifferentiation of the dorsal and ventral retinal pigment epithelium, defective optic cup periphery, and closure defects of the eyelid, as well as defective corneal morphogenesis. Thus, Porcn is required in both extraocular and neuroectodermal tissues to regulate distinct Wnt-dependent processes during morphogenesis of the posterior and anterior segments of the eye.


Subject(s)
Eye/embryology , Focal Dermal Hypoplasia/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Acyltransferases , Alleles , Animals , Disease Models, Animal , Eye/metabolism , Female , Genotype , Glycoproteins/metabolism , Hemizygote , In Situ Hybridization , Ligands , Male , Mice , Mice, Inbred C57BL , Mutation , Recombination, Genetic , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Wnt Proteins/metabolism
8.
Exp Eye Res ; 123: 141-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24060344

ABSTRACT

The retinal pigment epithelium (RPE) is a simple epithelium interposed between the neural retina and the choroid. Although only 1 cell-layer in thickness, the RPE is a virtual workhorse, acting in several capacities that are essential for visual function and preserving the structural and physiological integrities of neighboring tissues. Defects in RPE function, whether through chronic dysfunction or age-related decline, are associated with retinal degenerative diseases including age-related macular degeneration. As such, investigations are focused on developing techniques to replace RPE through stem cell-based methods, motivated primarily because of the seemingly limited regeneration or self-repair properties of mature RPE. Despite this, RPE cells have an unusual capacity to transdifferentiate into various cell types, with the particular fate choices being highly context-dependent. In this review, we describe recent findings elucidating the mechanisms and steps of RPE development and propose a developmental framework for understanding the apparent contradiction in the capacity for low self-repair versus high transdifferentiation.


Subject(s)
Homeostasis/physiology , Regeneration/physiology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Animals , Cell Transdifferentiation , Humans , Stem Cell Transplantation
9.
Dev Biol ; 371(1): 57-65, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22921921

ABSTRACT

Dorsal retinal fate is established early in eye development, via expression of spatially restricted dorsal-specific transcription factors in the optic vesicle; yet the events leading to initiation of dorsal fate are not clear. We hypothesized that induction of dorsal fate would require an extraocular signal arising from a neighboring tissue to pattern the prospective dorsal retina, however no such signal has been identified. We used the zebrafish embryo to determine the source, timing, and identity of the dorsal retina-inducing signal. Extensive cell movements occur during zebrafish optic vesicle morphogenesis, however the location of prospective dorsal cells within the early optic vesicle and their spatial relationship to early dorsal markers is currently unknown. Our mRNA expression and fate mapping analyses demonstrate that the dorsolateral optic vesicle is the earliest region to express dorsal specific markers, and cells from this domain contribute to the dorsal retinal pole at 24 hpf. We show that three bmp genes marking dorsal retina at 25 hpf are also expressed extraocularly before retinal patterning begins. We identified gdf6a as a dorsal initiation signal acting from the extraocular non-neural ectoderm during optic vesicle evagination. We find that bmp2b is involved in dorsal retina initiation, acting upstream of gdf6a. Together, this work has identified the nature and source of extraocular signals required to pattern the dorsal retina.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Ectoderm/physiology , Eye/embryology , Gene Expression Regulation, Developmental/physiology , Growth Differentiation Factor 6/metabolism , Morphogenesis/physiology , Retina/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Differentiation/physiology , DNA Primers/genetics , Ectoderm/metabolism , Gene Expression Regulation, Developmental/genetics , Genotype , In Situ Hybridization , Polymerase Chain Reaction , Pyrazoles , Pyrimidines , Retina/cytology , Zebrafish/genetics
10.
Curr Top Dev Biol ; 93: 61-84, 2010.
Article in English | MEDLINE | ID: mdl-20959163

ABSTRACT

Organogenesis of the eye is a multistep process that starts with the formation of optic vesicles followed by invagination of the distal domain of the vesicles and the overlying lens placode resulting in morphogenesis of the optic cup. The late optic vesicle becomes patterned into distinct ocular tissues: the neural retina, retinal pigment epithelium (RPE), and optic stalk. Multiple congenital eye disorders, including anophthalmia or microphthalmia, aniridia, coloboma, and retinal dysplasia, stem from disruptions in embryonic eye development. Thus, it is critical to understand the mechanisms that lead to initial specification and differentiation of ocular tissues. An accumulating number of studies demonstrate that a complex interplay between inductive signals provided by tissue-tissue interactions and cell-intrinsic factors is critical to ensuring proper specification of ocular tissues as well as maintenance of RPE cell fate. While several of the extrinsic and intrinsic determinants have been identified, we are just at the beginning in understanding how these signals are integrated. In addition, we know very little about the actual output of these interactions. In this chapter, we provide an update of the mechanisms controlling the early steps of eye development in vertebrates, with emphasis on optic vesicle evagination, specification of neural retina and RPE at the optic vesicle stage, the process of invagination during morphogenesis of the optic cup, and maintenance of the RPE cell fate.


Subject(s)
Eye/embryology , Morphogenesis/physiology , Animals , Eye/anatomy & histology , Gene Expression Regulation, Developmental , Lens, Crystalline/embryology , Retina/cytology , Retina/embryology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/embryology , Signal Transduction
11.
Invest Ophthalmol Vis Sci ; 51(10): 5328-35, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20463321

ABSTRACT

PURPOSE: Development of the retinal pigment epithelium (RPE) is controlled by intrinsic and extrinsic regulators including orthodenticle homeobox 2 (Otx2) and the Wnt/ß-catenin pathway, respectively. Otx2 and ß-catenin are necessary for the expression of the RPE key regulator microphthalmia-associated transcription factor (Mitf); however, neither factor is sufficient to promote Mitf expression in vivo. The study was conducted to determine whether Otx2 and ß-catenin act in a combinatorial manner and tested whether co-expression in the presumptive chick retina induces ectopic Mitf expression. METHODS: The sufficiency of Wnt/ß-catenin activation and/or Otx2 expression to induce RPE-specific gene expression was examined in chick optic vesicle explant cultures or in the presumptive neural retina using in ovo-electroporation. Luciferase assays were used to examine the transactivation potentials of Otx2 and ß-catenin on the Mitf-D enhancer and autoregulation of the Mitf-D and Otx2T0 enhancers. RESULTS: In optic vesicles explant cultures, RPE-specific gene expression was activated by lithium chloride, a Wnt/ß-catenin agonist. However, in vivo, Mitf was induced only in the presumptive retina if both ß-catenin and Otx2 are co-expressed. Furthermore, both Mitf and Otx2 can autoregulate their own enhancers in vitro. CONCLUSIONS: The present study provides evidence that ß-catenin and Otx2 are sufficient, at least in part, to convert retinal progenitor cells into presumptive RPE cells expressing Mitf. Otx2 may act as a competence factor that allows RPE specification in concert with additional RPE-promoting factors such as ß-catenin.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Microphthalmia-Associated Transcription Factor/genetics , Otx Transcription Factors/genetics , Retina/embryology , Retinal Pigment Epithelium/cytology , Transfection , beta Catenin/genetics , Animals , Cell Count , Cell Differentiation , Cells, Cultured , Chick Embryo , Electroporation , Fluorescent Antibody Technique, Indirect , Microscopy, Confocal , Plasmids , Retinal Neurons/cytology , Retinal Pigment Epithelium/metabolism , Transcriptional Activation , Wnt Proteins/genetics
12.
Hum Mol Genet ; 19(9): 1791-804, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20150232

ABSTRACT

Appropriate development of the retina and optic nerve requires that the forebrain-derived optic neuroepithelium undergoes a precisely coordinated sequence of patterning and morphogenetic events, processes which are highly influenced by signals from adjacent tissues. Our previous work has suggested that transcription factor activating protein-2 alpha (AP-2alpha; Tcfap2a) has a non-cell autonomous role in optic cup (OC) development; however, it remained unclear how OC abnormalities in AP-2alpha knockout (KO) mice arise at the morphological and molecular level. In this study, we show that patterning and morphogenetic defects in the AP-2alpha KO optic neuroepithelium begin at the optic vesicle stage. During subsequent OC formation, ectopic neural retina and optic stalk-like tissue replaced regions of retinal pigment epithelium. AP-2alpha KO eyes also displayed coloboma in the ventral retina, and a rare phenotype in which the optic stalk completely failed to extend, causing the OCs to be drawn inward to the midline. We detected evidence of increased sonic hedgehog signaling in the AP-2alpha KO forebrain neuroepithelium, which likely contributed to multiple aspects of the ocular phenotype, including expansion of PAX2-positive optic stalk-like tissue into the OC. Our data suggest that loss of AP-2alpha in multiple tissues in the craniofacial region leads to severe OC and optic stalk abnormalities by disturbing the tissue-tissue interactions required for ocular development. In view of recent data showing that mutations in human TFAP2A result in similar eye defects, the current findings demonstrate that AP-2alpha KO mice provide a valuable model for human ocular disease.


Subject(s)
Disease Models, Animal , Eye Abnormalities/metabolism , Gene Expression Regulation, Developmental/physiology , Morphogenesis/genetics , Optic Nerve/embryology , Retina/embryology , Transcription Factor AP-2/genetics , Animals , DNA Primers/genetics , Eye Abnormalities/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/metabolism , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mice, Knockout , Morphogenesis/physiology , Polymerase Chain Reaction , Prosencephalon/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor AP-2/metabolism
13.
Development ; 136(15): 2505-10, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19553286

ABSTRACT

The retinal pigment epithelium (RPE) consists of a monolayer of cuboidal, pigmented cells that is located between the retina and the choroid. The RPE is vital for growth and function of the vertebrate eye and improper development results in congenital defects, such as microphthalmia or anophthalmia, or a change of cell fate into neural retina called transdifferentiation. The transcription factors microphthalmia-associated transcription factor (Mitf) and orthodenticle homolog 2 (Otx2) are crucial for RPE development and function; however, very little is known about their regulation. Here, by using a Wnt-responsive reporter, we show that the Wnt/beta-catenin pathway is activated in the differentiating mouse RPE. Cre-mediated, RPE-specific disruption of beta-catenin after the onset of RPE specification causes severe defects, resulting in microphthalmia with coloboma, disturbed lamination, and mislocalization of adherens junction proteins. Upon beta-catenin deletion, the RPE transforms into a multilayered tissue in which the expression of Mitf and Otx2 is downregulated, while retina-specific gene expression is induced, which results in the transdifferentiation of RPE into retina. Chromatin immunoprecipitation (ChIP) and luciferase assays indicate that beta-catenin binds near to and activates potential TCF/LEF sites in the Mitf and Otx2 enhancers. We conclude that Wnt/beta-catenin signaling is required for differentiation of the RPE by directly regulating the expression of Mitf and Otx2. Our study is the first to show that an extracellular signaling pathway directly regulates the expression of RPE-specific genes such as Mitf and Otx2, and elucidates a new role for the Wnt/beta-catenin pathway in organ formation and development.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Microphthalmia-Associated Transcription Factor/genetics , Otx Transcription Factors/genetics , Retinal Pigment Epithelium/cytology , beta Catenin/metabolism , Adherens Junctions/metabolism , Animals , Binding Sites , Cell Adhesion , Cell Transdifferentiation , Enhancer Elements, Genetic/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Membrane Proteins/metabolism , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Mutation/genetics , Neurons/cytology , Organ Specificity , Otx Transcription Factors/metabolism , Protein Binding , Protein Transport , Retinal Pigment Epithelium/abnormalities , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/pathology , beta Catenin/deficiency
14.
Invest Ophthalmol Vis Sci ; 50(1): 432-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18599572

ABSTRACT

PURPOSE: High mobility group (HMG) transcription factors of the T-cell-specific transcription factor/lymphoid enhancer binding factor (TCF/LEF) family are a class of intrinsic regulators that are dynamically expressed in the embryonic mouse retina. Activation of TCF/LEFs is a hallmark of the Wnt/beta-catenin pathway; however, the requirement for Wnt/beta-catenin and noncanonical Wnt signaling during mammalian retinal development remains unclear. The goal of the study was to characterize more fully a TCF/LEF-responsive retinal progenitor population in the mouse embryo and to correlate this with Wnt/beta-catenin signaling. METHODS: TCF/LEF activation was analyzed in the TOPgal (TCF optimal promoter) reporter mouse at embryonic ages and compared to Axin2 mRNA expression, an endogenous readout of Wnt/beta-catenin signaling. Reporter expression was also examined in embryos with a retina-specific deletion of the beta-catenin gene (Ctnnb1), using Six3-Cre transgenic mice. Finally, the extent to which TOPgal cells coexpress cell cycle proteins, basic helix-loop-helix (bHLH) transcription factors, and other retinal cell markers was tested by double immunohistochemistry. RESULTS: TOPgal reporter activation occurred transiently in a subpopulation of embryonic retinal progenitor cells. Axin2 was not expressed in the central retina, and TOPgal reporter expression persisted in the absence of beta-catenin. Although a proportion of TOPgal-labeled cells were proliferative, most coexpressed the cyclin-dependent kinase inhibitor p27/Kip1. CONCLUSIONS: TOPgal cells give rise to the four earliest cell types: ganglion, amacrine, horizontal, and photoreceptor. TCF/LEF activation in the central retina does not correlate with Wnt/beta-catenin signaling, pointing to an alternate role for this transcription factor family during retinal development.


Subject(s)
Embryonic Stem Cells/metabolism , Retina/embryology , TCF Transcription Factors/metabolism , Animals , Axin Protein , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/genetics , Fluorescent Antibody Technique, Indirect , In Situ Hybridization , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
15.
Invest Ophthalmol Vis Sci ; 49(12): 5561-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18791178

ABSTRACT

PURPOSE: Frizzled-5 (Fzd5) is expressed in the developing retina of multiple species and appears to play species-specific roles during eye development. The present study analyzed the effects of tissue-specific deletion of Fzd5 on mammalian eye development. METHODS: To generate Fzd5 conditional knockout (CKO) mice, Fzd5(+/-) mice carrying the Six3-Cre transgene were crossed with Fzd5(LoxP/LoxP) mice. To determine which cell lineages in the eye had Cre recombinase activity, Six3-Cre transgenic mice were crossed with ROSA-26 reporter mice, and lacZ activity was assayed. Histologic analysis, immunofluorescence, and TUNEL labeling were performed from embryonic day (E)12.5 to postnatal stages to analyze vascularization, cell proliferation, retinal organization, and apoptosis. RESULTS: On conditional disruption of Fzd5 specifically in the retina, but not in vitreous hyaloid vasculature (VHV), an abnormal accumulation consisting of pericytes and endothelial cells was observed in the vitreous as early as E12.5. The abundant retrolental cells persisted into postnatal stages and appeared as a pigmented intravitreal mass. In Fzd5 CKO mice there was failure of normal apoptosis of the VHV, and cells in the persistent VHV were maintained in the cell cycle up to postnatal day 23. Moreover, morphogenesis of the retina adjacent to the vasculature was disrupted, leading to retinal folds, detachment, and abnormal lamination. This phenotype is similar to that of human eye disease persistent hyperplastic primary vitreous (PHPV). CONCLUSIONS: Selective loss of Fzd5 in the retina results in PHPV and retinal defects through an apparently cell-nonautonomous effect, revealing a potential requirement for retina-derived signals in regulating the development of the VHV.


Subject(s)
Frizzled Receptors/physiology , Persistent Hyperplastic Primary Vitreous/embryology , Receptors, G-Protein-Coupled/physiology , Retina/embryology , Vitreous Body/blood supply , Animals , Apoptosis , Cell Proliferation , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental/physiology , Gene Silencing , Genotype , In Situ Hybridization , In Situ Nick-End Labeling , Integrases/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Persistent Hyperplastic Primary Vitreous/pathology , Polymerase Chain Reaction , Retina/pathology , Retinal Dysplasia/embryology , Retinal Dysplasia/pathology , Retinal Ganglion Cells/pathology
16.
Dev Dyn ; 237(6): 1614-26, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18489003

ABSTRACT

Recent studies revealed that the Wnt receptor Frizzled-5 (Fzd5) is required for eye and retina development in zebrafish and Xenopus, however, its role during mammalian eye development is unknown. In the mouse embryo, Fzd5 is prominently expressed in the pituitary, distal optic vesicle, and optic stalk, then later in the progenitor zone of the developing retina. To elucidate the role of Fzd5 during eye development, we analyzed embryos with a germline disruption of the Fzd5 gene at E10.25, just before embryos die due to defects in yolk sac angiogenesis. We observed severe defects in optic cup morphogenesis and lens development. However, in embryos with conditional inactivation of Fzd5 using Six3-Cre, we observed no obvious early eye defects. Analysis of Axin2 mRNA expression and TCF/LEF-responsive reporter activation demonstrate that Fzd5 does not regulate the Wnt/beta-catenin pathway in the eye. Thus, the function of Fzd5 during eye development appears to be species-dependent.


Subject(s)
Eye/embryology , Frizzled Receptors/biosynthesis , Frizzled Receptors/physiology , Gene Expression Regulation, Developmental , Neurons/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/physiology , Animals , Cell Death , Cell Proliferation , Lens, Crystalline/embryology , Mice , Mice, Transgenic , Mutation , Optic Nerve/embryology , Pituitary Gland/metabolism , Retina/embryology , Time Factors
17.
Organogenesis ; 4(2): 60-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-19122781

ABSTRACT

The vertebrate eye consists of multiple tissues with distinct embryonic origins. To ensure formation of the eye as a functional organ, development of ocular tissues must be precisely coordinated. Besides intrinsic regulators, several extracellular pathways have been shown to participate in controlling critical steps during eye development. Many components of Wnt/Frizzled signaling pathways are expressed in developing ocular tissues, and substantial progress has been made in the past few years in understanding their function during vertebrate eye development. Here, I summarize recent work using functional experiments to elucidate the roles of Wnt/Frizzled pathways during development of ocular tissues in different vertebrates.

18.
Invest Ophthalmol Vis Sci ; 45(2): 655-61, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14744911

ABSTRACT

PURPOSE: Application of ciliary neurotrophic factor (CNTF) can rescue mature photoreceptors from lesion-induced and hereditary degeneration. In the chick retina, expression of the CNTF receptor is present in a subpopulation of photoreceptor cells. The present study was undertaken to identify the CNTF receptor-expressing photoreceptors and to describe the subcellular localization of the receptor protein. METHODS: The localization of the CNTF receptor was analyzed by light and electron microscopic immunocytochemistry in chick retinal wholemount preparations, with an antibody for CNTF receptor alpha (CNTFRalpha). Immunoreactive cells were identified by double labeling with immunocytochemical markers for photoreceptor subpopulations. RESULTS: The CNTFRalpha antibody labeled evenly distributed outer segments (OS) of a photoreceptor subpopulation. CNTFRalpha-positive OS were associated with oil droplets of uniform size. Receptor immunoreactivity did not colocalize with markers for rods and red-green cones. Complete overlap was found after double labeling with the antibody CERN 933, which recognizes violet-sensitive cones in the chick retina. Ultrastructurally, the CNTFRalpha-immunoreactive OS showed rodlike properties: an elongated shape and stacks of membrane discs separated from the plasma membrane. Immunoreactivity was completely restricted to the plasma membrane of the OS and the inner membrane sheet of the photoreceptor calices present in avian retinas. CONCLUSIONS: CNTFRalpha expression identifies a unique type of photoreceptors in the avian retina which does not fit into the classic morphologic definition of rods and cones. The specific expression in violet-sensitive photoreceptors suggests that CNTF may have a neuroprotective role related to the specific function of these cells.


Subject(s)
Chickens/anatomy & histology , Receptor, Ciliary Neurotrophic Factor/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Animals , Chick Embryo , Fluorescent Antibody Technique, Indirect , In Situ Hybridization , Light , Microscopy, Immunoelectron , Retinal Cone Photoreceptor Cells/embryology
19.
Gene Expr Patterns ; 3(5): 659-62, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12972002

ABSTRACT

Frizzleds are transmembrane receptors that can transduce signals dependent upon binding of Wnts, a large family of secreted glycoproteins homologous to the Drosophila wingless (wg) gene product and critical for a wide variety of normal and pathological developmental processes. In the nervous system, Wnts and Frizzleds play an important role in anterior-posterior patterning, cell fate decisions, proliferation, and synaptogenesis. However, little is known about the role of Frizzled signaling in the developing eye. We isolated cDNAs for ten chick Frizzleds and analyzed the spatial and temporal expression patterns during eye development in the chick embryo. Frizzled-1 to -9 are specifically expressed in the eye at various stages of development and show a complex and partially overlapping pattern of expression.


Subject(s)
Chick Embryo , Eye/embryology , Proteins/genetics , Amino Acid Sequence , Animals , Eye/metabolism , Frizzled Receptors , Gene Expression Profiling , Gene Expression Regulation, Developmental , Molecular Sequence Data , Proteins/metabolism , Signal Transduction
20.
J Comp Neurol ; 461(1): 111-22, 2003 Jun 16.
Article in English | MEDLINE | ID: mdl-12722108

ABSTRACT

Ciliary neurotrophic factor (CNTF) promotes the survival and differentiation of various neuronal and glial cell populations in the nervous system of vertebrates. In mammals, the ligand-binding alpha-subunit of the CNTF receptor (CNTFRalpha) is expressed in a variety of neuronal populations, including all CNTF-responsive cells. Previous studies suggested that functional differences in the CNTF/CNTF receptor system between chicks and mammals exist. The purpose of the present study was to examine the temporal and spatial expression pattern of the chick CNTFRalpha protein during CNS development. Receptor expression was detectable by immunoblotting in all CNS areas tested but showed area-specific developmental regulation. Interestingly, two variants of CNTFRalpha, 69 and 65 kD, were identified by immunoblotting with a shift from the higher to the lower molecular mass species occurring during development. Immunoreactivity for CNTFRalpha protein was preferentially observed in neuropil and white matter structures of the developing CNS while neuronal somata generally appeared unlabeled. For example, expression was observed in the olfactory system, in the telencephalon, in parts of the somatosensory system, in components of the tectofugal pathway, in the cerebellum, and in auditory brainstem nuclei. Fiber tracts that exhibit CNTFRalpha immunoreactivity were the lateral forebrain bundle, occipitomesencephalic tract, quintofrontal tract, and vestibular nerve. Our study identifies potential new targets of a chick CNTF-related molecule and reveals significant regional differences of CNTFRalpha protein expression between chick and mammals. These results suggest that the CNTF receptor performs distinct developmental functions in different animals.


Subject(s)
Brain/metabolism , Chick Embryo/metabolism , Gene Expression Regulation, Developmental/physiology , Receptor, Ciliary Neurotrophic Factor/metabolism , Animals , Brain/embryology , Immunoblotting , Immunohistochemistry , Organogenesis/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...