Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
J Dev Biol ; 10(4)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36412639

ABSTRACT

The first event of differentiation and morphogenesis in the optic vesicle (OV) is specification of the neural retina (NR) and retinal pigment epithelium (RPE), separating the inner and outer layers of the optic cup, respectively. Here, we focus on a basic helix-loop-helix gene, BHLHE40, which has been shown to be expressed by the developing RPE in mice and zebrafish. Firstly, we examined the expression pattern of BHLHE40 in the developing chicken eye primordia by in situ hybridization. Secondly, BHLHE40 overexpression was performed with in ovo electroporation and its effects on optic cup morphology and expression of NR and RPE marker genes were examined. Thirdly, we examined the expression pattern of BHLHE40 in LHX1-overexpressed optic cup. BHLHE40 expression emerged in a subset of cells of the OV at Hamburger and Hamilton stage 14 and became confined to the outer layer of the OV and the ciliary marginal zone of the retina by stage 17. BHLHE40 overexpression in the prospective NR resulted in ectopic induction of OTX2 and repression of VSX2. Conversely, BHLHE40 was repressed in the second NR after LHX1 overexpression. These results suggest that emergence of BHLHE40 expression in the OV is involved in initial RPE specification and that BHLHE40 plays a role in separation of the early OV domains by maintaining OTX2 expression and antagonizing an NR developmental program.

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.
Biol Open ; 10(10)2021 10 15.
Article in English | MEDLINE | ID: mdl-34590124

ABSTRACT

The morphogenesis of the vertebrate eye consists of a complex choreography of cell movements, tightly coupled to axial regionalization and cell type specification processes. Disturbances in these events can lead to developmental defects and blindness. Here, we have deciphered the sequence of defective events leading to coloboma in the embryonic eye of the blind cavefish of the species Astyanax mexicanus. Using comparative live imaging on targeted enhancer-trap Zic1:hsp70:GFP reporter lines of both the normal, river-dwelling morph and the cave morph of the species, we identified defects in migratory cell behaviours during evagination that participate in the reduced optic vesicle size in cavefish, without proliferation defect. Further, impaired optic cup invagination shifts the relative position of the lens and contributes to coloboma in cavefish. Based on these results, we propose a developmental scenario to explain the cavefish phenotype and discuss developmental constraints to morphological evolution. The cavefish eye appears as an outstanding natural mutant model to study molecular and cellular processes involved in optic region morphogenesis.


Subject(s)
Blindness/genetics , Eye/growth & development , Fishes/growth & development , Morphogenesis/genetics , Animals , Evolution, Molecular
4.
J Neurol Surg B Skull Base ; 82(1): 2-6, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33777615

ABSTRACT

The orbit houses and protects the ocular globe and the supporting structures, and occupies a strategic position below the anterior skull base and adjacent to the paranasal sinuses. Its embryologic origins are inextricably intertwined with those of the central nervous system, skull base, and face. Although the orbit contains important contributions from four germ cell layers (surface ectoderm, neuroectoderm, neural crest, and mesoderm), a significant majority originate from the neural crest cells. The bones of the orbit, face, and anterior cranial vault are mostly neural crest in origin. The majority of the bones of the skull base are formed through endochondral ossification, whereas the cranial vault is formed through intramembranous ossification. Familiarity with the embryology and fetal development of the orbit can aid in understanding its anatomy, as well as many developmental anomalies and pathologic conditions that affect the orbit.

5.
Biochem Biophys Res Commun ; 543: 80-86, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33548738

ABSTRACT

The optic vesicle in the developing embryonic eye contains a multitude of neuroepithelial progenitors that subsequently differentiate into functionally distinct domains of the optic cup, such as the neural retina, pigment epithelium, and optic stalk. To investigate cell-type diversity across early optic vesicles before regionalization of the optic cup, we performed single-cell RNA-sequencing (scRNA-seq) using 7989 cells from the presumptive eye area in mouse embryos at the 12-26-somite stages at five developmental time points. We demonstrated the presence of seven optic vesicle populations. Moreover, the four populations of retinal progenitor cells could be classified according to their stage-dependent time point, and these cells exhibited altered expression of several structural and metabolic key genes, such as Col9a1 and Ckb, just before regionalization of the optic cup. From these data, we provide the first report on stage-dependent transcriptional profiles during initial retinal specification at single-cell resolution and highlight the unexpected developmental heterogeneity of the murine optic vesicle structure.


Subject(s)
Embryo, Nonmammalian/metabolism , Eye/growth & development , Eye/metabolism , Retina/metabolism , Single-Cell Analysis/methods , Stem Cells/metabolism , Animals , Female , Mice , Mice, Inbred ICR , Retina/cytology , Signal Transduction , Stem Cells/cytology , Transcriptome
6.
Dev Dyn ; 250(6): 807-821, 2021 06.
Article in English | MEDLINE | ID: mdl-32864847

ABSTRACT

BACKGROUND: Vertebrate eye formation requires coordinated inductive interactions between different embryonic tissue layers, first described in amphibians. A network of transcription factors and signaling molecules controls these steps, with mutations causing severe ocular, neuronal, and craniofacial defects. In eyeless mutant axolotls, eye morphogenesis arrests at the optic vesicle stage, before lens induction, and development of ventral forebrain structures is disrupted. RESULTS: We identified a 5-bp deletion in the rax (retina and anterior neural fold homeobox) gene, which was tightly linked to the recessive eyeless (e) axolotl locus in an F2 cross. This frameshift mutation, in exon 2, truncates RAX protein within the homeodomain (P154fs35X). Quantitative RNA analysis shows that mutant and wild-type rax transcripts are equally abundant in E/e embryos. Translation appears to initiate from dual start codons, via leaky ribosome scanning, a conserved feature among gnathostome RAX proteins. Previous data show rax is expressed in the optic vesicle and diencephalon, deeply conserved among metazoans, and required for eye formation in other species. CONCLUSION: The eyeless axolotl mutation is a null allele in the rax homeobox gene, with primary defects in neural ectoderm, including the retinal and hypothalamic primordia.


Subject(s)
Ambystoma mexicanum/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , Ambystoma mexicanum/metabolism , Animals , Embryonic Development/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription Factors/metabolism
7.
Dev Biol ; 462(2): 119-128, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32169553

ABSTRACT

Arl13b is a gene known to regulate ciliogenesis. Functional alterations in this gene's activity have been associated with Joubert syndrome. We found that in Arl13 null mouse embryos the orientation of the optic cup is inverted, such that the lens is abnormally surrounded by an inverted optic cup whose retina pigmented epithelium is oddly facing the surface ectoderm. Loss of Arl13b leads to the disruption of optic vesicle's patterning and expansion of ventral fates. We show that this phenotype is consequence of miss-regulation of Sonic hedgehog (Shh) signaling and demonstrate that the Arl13b-/- eye phenotype can be rescued by deletion of Gli2, a downstream effector of the Shh pathway. This work identified an unexpected role of primary cilia during the morphogenetic movements required for the formation of the eye.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cilia/metabolism , Eye/embryology , ADP-Ribosylation Factors/genetics , Animals , Body Patterning/genetics , Bone Morphogenetic Protein 4/metabolism , Cilia/genetics , Embryonic Development , Eye/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Male , Mice , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organogenesis , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Signal Transduction/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , Homeobox Protein SIX3
8.
Neuroscientist ; 26(2): 185-196, 2020 04.
Article in English | MEDLINE | ID: mdl-31509088

ABSTRACT

The primordium of the vertebrate eye is composed of a pseudostratified and apparently homogeneous neuroepithelium, which folds inward to generate a bilayered optic cup. During these early morphogenetic events, the optic vesicle is patterned along three different axes-proximo-distal, dorso-ventral, and naso-temporal-and three major domains: the neural retina, the retinal pigment epithelium (RPE), and the optic stalk. These fundamental steps that enable the subsequent development of a functional eye, entail the precise coordination among genetic programs. These programs are driven by the interplay of signaling pathways and transcription factors, which progressively dictate how each tissue should evolve. Here, we discuss the contribution of the Hh, Wnt, FGF, and BMP signaling pathways to the early patterning of the retina. Comparative studies in different vertebrate species have shown that their morphogenetic activity is repetitively used to orchestrate the progressive specification of the eye with evolutionary conserved mechanisms that have been adapted to match the specific need of a given species.


Subject(s)
Image Processing, Computer-Assisted , Morphogenesis/physiology , Retina/pathology , Retina/physiopathology , Signal Transduction/physiology , Animals , Hedgehog Proteins/metabolism , Humans , Retina/metabolism , Signal Transduction/genetics , Vertebrates/metabolism
9.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-733634

ABSTRACT

Objective To investigate the regulation effects of Ngn2 gene transfection on retinal neuron differnetion in three-dimentional optic vesicle (OV) of mice.Methods OV was cultured in vitro using mouse induced pluripotent stem cells (iPSC) under specific conditions.During OV culture,it was transfected multiple times by lentivirus-mediated Ngn2 gene and then it was induced after maturation.The cells were specificly differentiated toward retinal nerve cells in OV.Using the green fluorescent protein (EGFP) gene as control,the differentiation of retinal nerve cells in OV was detected by immunohistochemistry.Reverse transcription PCR and Western blot were used to quantitatively detect the expressions of retinal neuron-specific proteins Pax6,Islet1 and Brn3b.Results The mouse iPS-derived OV was successfully cultured.The number of neural cells in the OV transfected with the Ngn2 gene was increased by the lentiviral-mediated lentivirus.The expressions of PAX6,Islet1 and Brn3b in the Ngn2 transfection group were significantly higher at the gene and protein levels than those in the control group,with significant differences between the two groups (P<0.05).Conclusions The Ngn2 gene can effectively increase the number of retinal neuron differentiation in OV and make in vitro cultured OV more mature and form a more perfect retinal cell neural circuit.

10.
Front Cell Neurosci ; 12: 42, 2018.
Article in English | MEDLINE | ID: mdl-29515375

ABSTRACT

Coloboma is a defect in the morphogenesis of the eye that is a consequence of failure of choroid fissure fusion. It is among the most common congenital defects in humans and can significantly impact vision. However, very little is known about the cellular mechanisms that regulate choroid fissure closure. Using high-resolution confocal imaging of the zebrafish optic cup, we find that apico-basal polarity is re-modeled in cells lining the fissure in proximal to distal and inner to outer gradients during fusion. This process is accompanied by cell proliferation, displacement of vasculature, and contact between cells lining the choroid fissure and periocular mesenchyme (POM). To investigate the role of POM cells in closure of the fissure, we transplanted optic vesicles onto the yolk, allowing them to develop in a situation where they are depleted of POM. The choroid fissure forms normally in ectopic eyes but fusion fails in this condition, despite timely apposition of the nasal and temporal lips of the retina. This study resolves some of the cell behaviors underlying choroid fissure fusion and supports a role for POM in choroid fissure fusion.

11.
Prog Biophys Mol Biol ; 137: 25-36, 2018 09.
Article in English | MEDLINE | ID: mdl-29432780

ABSTRACT

In the vertebrate embryo, the eyes develop from optic vesicles that grow laterally outward from the brain tube and contact the overlying surface ectoderm. Within the region of contact, each optic vesicle and the surface ectoderm thicken to form placodes, which then invaginate to create the optic cup and lens pit, respectively. Eventually, the optic cup becomes the retina, while the lens pit closes to form the lens vesicle. Here, we review current hypotheses for the physical mechanisms that create these structures and present novel three-dimensional computer (finite-element) models to illustrate the plausibility and limitations of these hypotheses. Taken together, experimental and numerical results suggest that the driving forces for early eye morphogenesis are generated mainly by differential growth, actomyosin contraction, and regional apoptosis, with morphology mediated by physical constraints provided by adjacent tissues and extracellular matrix. While these studies offer new insight into the mechanics of eye development, future work is needed to better understand how these mechanisms are regulated to precisely control the shape of the eye.


Subject(s)
Eye/growth & development , Mechanical Phenomena , Animals , Biomechanical Phenomena , Eye/anatomy & histology , Humans , Lens, Crystalline/anatomy & histology , Lens, Crystalline/growth & development
12.
Dev Biol ; 428(1): 88-100, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28576690

ABSTRACT

Complement components have been implicated in a wide variety of functions including neurogenesis, proliferation, cell migration, differentiation, cancer, and more recently early development and regeneration. Following our initial observations indicating that C3a/C3aR signaling induces chick retina regeneration, we analyzed its role in chick eye morphogenesis. During eye development, the optic vesicle (OV) invaginates to generate a bilayer optic cup (OC) that gives rise to the retinal pigmented epithelium (RPE) and neural retina. We show by immunofluorescence staining that C3 and the receptor for C3a (the cleaved and active form of C3), C3aR, are present in chick embryos during eye morphogenesis in the OV and OC. Interestingly, C3aR is mainly localized in the nuclear compartment at the OC stage. Loss of function studies at the OV stage using morpholinos or a blocking antibody targeting the C3aR (anti-C3aR Ab), causes eye defects such as microphthalmia and defects in the ventral portion of the eye that result in coloboma. Such defects were not observed when C3aR was disrupted at the OC stage. Histological analysis demonstrated that microphthalmic eyes were unable to generate a normal optic stalk or a closed OC. The dorsal/ventral patterning defects were accompanied by an expansion of the ventral markers Pax2, cVax and retinoic acid synthesizing enzyme raldh-3 (aldh1a3) domains, an absence of the dorsal expression of Tbx5 and raldh-1 (aldh1a1) and a re-specification of the ventral RPE to neuroepithelium. In addition, the eyes showed overall decreased expression of Gli1 and a change in distribution of nuclear ß-catenin, suggesting that Shh and Wnt pathways have been affected. Finally, we observed prominent cell death along with a decrease in proliferating cells, indicating that both processes contribute to the microphthalmic phenotype. Together our results show that C3aR is necessary for the proper morphogenesis of the OC. This is the first report implicating C3aR in eye development, revealing an unsuspected hitherto regulator for proper chick eye morphogenesis.


Subject(s)
Body Patterning/physiology , Complement C3a/metabolism , Gene Expression Regulation, Developmental , Receptors, Complement/metabolism , Retinal Pigment Epithelium/embryology , Aldehyde Dehydrogenase/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Chick Embryo , Hedgehog Proteins/metabolism , Microphthalmos/embryology , Morphogenesis/physiology , PAX2 Transcription Factor/metabolism , Receptors, Complement/genetics , Retinal Dehydrogenase/metabolism , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway/physiology , Zinc Finger Protein GLI1/biosynthesis , beta Catenin/metabolism
13.
Dev Growth Differ ; 58(9): 741-749, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27921294

ABSTRACT

The chick embryonic eye is an excellent model for the study of vertebrate organogenesis. Key events in eye development involve thickening, invagination and cytodifferentiation of the lens primordium. While these events occur successively at different developmental stages, the extent to which these events are temporally related is largely unknown. Here we show that the lens invagination is highly sensitive to temperature. Lowering of incubation temperature to 29°C at embryonic day 2 delayed the onset of invagination of the lens, but not thickening and cytodifferentiation, leading to abnormal protrusion of the eye. The temperature shift also delayed the inward bending of the underlying retinal primordium, even in the absence of the lens. Taken together, our results suggest that lens invagination is initiated independently of thickening and cytodifferentiation, possibly by mechanisms associated with morphogenesis of the primordial retina.


Subject(s)
Cell Differentiation , Cold Temperature , Lens, Crystalline/embryology , Organogenesis , Retina/embryology , Animals , Chick Embryo , Lens, Crystalline/cytology , Retina/cytology
14.
Mech Dev ; 141: 90-99, 2016 08.
Article in English | MEDLINE | ID: mdl-27151576

ABSTRACT

The eyes are subdivided from the rostral diencephalon in early development. How the neuroectoderm regulates this subdivision, however, is largely unknown. Taking advantage of embryonic stem cell (ESC) culture using a Rax reporter line to monitor rostral diencephalon formation, we found that ESC-derived tissues at day 7 grown in Glasgow Minimum Expression Media (GMEM) containing knockout serum replacement (KSR) exhibited higher levels of expression of axin2, a Wnt target gene, than those grown in chemically defined medium (CDM). Surprisingly, Wnt agonist facilitated eye field-like tissue specification in CDM. In contrast, the addition of Wnt antagonist diminished eye field tissue formation in GMEM+KSR. Furthermore, the morphological formation of the eye tissue anlage, including the optic vesicle, was accompanied by Wnt signaling activation. Additionally, using CDM culture, we developed an efficient method for generating Rax+/Chx10+ retinal progenitors, which could become fully stratified retina. Here we provide a new avenue for exploring the mechanisms of eye field specification in vitro.


Subject(s)
Axin Protein/genetics , Embryonic Development/genetics , Embryonic Stem Cells/metabolism , Eye/growth & development , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Diencephalon/growth & development , Eye/embryology , Eye/metabolism , Eye Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Intralaminar Thalamic Nuclei/growth & development , Mice , Retina/growth & development , Retina/metabolism , Transcription Factors/genetics , Wnt Signaling Pathway/drug effects
15.
Curr Eye Res ; 41(4): 558-68, 2016 04.
Article in English | MEDLINE | ID: mdl-25880804

ABSTRACT

PURPOSE: To establish a practical research tool for studying the pathogenesis of retinal ganglion cell (RGC) diseases, we optimized culture procedures to induce neurite outgrowth from three-dimensional self-organizing optic vesicles (3D-retinas) differentiated in vitro from mouse and human embryonic stem cells (ESCs). MATERIALS AND METHODS: The developing 3D-retinas isolated at various time points were placed on Matrigel-coated plates and cultured in media on the basis of the 3D-retinal culture or the retinal organotypic culture protocol. The number, length, and morphology of the neurites in each culture condition were compared. RESULTS: First, we confirmed that Venus-positive cells were double-labeled with a RGC marker, Brn3a, in the 3D-retina differentiated from Fstl4::Venus mouse ESCs, indicating specific RGC-subtype differentiation. Second, Venus-positive neurites grown from these RGC subsets were positive for beta-III tubulin and SMI312 by immunohistochemistry. Enhanced neurite outgrowth was observed in the B27-supplemented Neurobasal-A medium on Matrigel-coated plates from the optic vesicles isolated after 14 days of differentiation from mouse ESCs. For the differentiated RGCs from human ESCs, we obtained neurite extension of >4 mm by modifying Matrigel coating and the culture medium from the mouse RGC culture. CONCLUSION: We successfully optimized the culture conditions to enhance lengthy and high-frequency neurite outgrowth in mouse and human models. The procedure would be useful for not only developmental studies of RGCs, including maintenance and projection, but also clinical, pathological, and pharmacological studies of human RGC diseases.


Subject(s)
Human Embryonic Stem Cells/cytology , Nerve Degeneration/therapy , Neurites/pathology , Neuronal Outgrowth/physiology , Retinal Ganglion Cells/pathology , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Humans , Imaging, Three-Dimensional , Immunohistochemistry , Mice , Mice, Knockout , Nerve Degeneration/pathology
16.
Curr Protoc Stem Cell Biol ; 32: 1H.8.1-1H.8.20, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25640818

ABSTRACT

The protocol outlined below is used to differentiate human pluripotent stem cells (hPSCs) into retinal cell types through a process that faithfully recapitulates the stepwise progression observed in vivo. From pluripotency, cells are differentiated to a primitive anterior neural fate, followed by progression into two distinct populations of retinal progenitors and forebrain progenitors, each of which can be manually separated and purified. The hPSC-derived retinal progenitors are found to self-organize into three-dimensional optic vesicle-like structures, with each aggregate possessing the ability to differentiate into all major retinal cell types. The ability to faithfully recapitulate the stepwise in vivo development in a three-dimensional cell culture system allows for the study of mechanisms underlying human retinogenesis. Furthermore, this methodology allows for the study of retinal dysfunction and disease modeling using patient-derived cells, as well as high-throughput pharmacological screening and eventually patient-specific therapies.


Subject(s)
Cell Culture Techniques/methods , Optic Disk/cytology , Pluripotent Stem Cells/cytology , Retina/cytology , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryoid Bodies/cytology , Humans , Neuroepithelial Cells/cytology , Retinal Pigment Epithelium/cytology , Spheroids, Cellular/cytology , Stem Cells/cytology
17.
Elife ; 42015 Feb 24.
Article in English | MEDLINE | ID: mdl-25719386

ABSTRACT

The hemispheric, bi-layered optic cup forms from an oval optic vesicle during early vertebrate eye development through major morphological transformations. The overall basal surface, facing the developing lens, is increasing, while, at the same time, the space basally occupied by individual cells is decreasing. This cannot be explained by the classical view of eye development. Using zebrafish (Danio rerio) as a model, we show that the lens-averted epithelium functions as a reservoir that contributes to the growing neuroretina through epithelial flow around the distal rims of the optic cup. We propose that this flow couples morphogenesis and retinal determination. Our 4D data indicate that future stem cells flow from their origin in the lens-averted domain of the optic vesicle to their destination in the ciliary marginal zone. BMP-mediated inhibition of the flow results in ectopic neuroretina in the RPE domain. Ultimately the ventral fissure fails to close resulting in coloboma.


Subject(s)
Bone Morphogenetic Proteins/physiology , Eye/growth & development , Morphogenesis , Optic Disk/physiology , Animals , Epithelium/physiology , Zebrafish
18.
Dev Dyn ; 244(3): 266-76, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25329498

ABSTRACT

BACKGROUND: During development of the vertebrate eye, optic tissue is progressively compartmentalized into functionally distinct tissues. From the central to the peripheral optic cup, the original optic neuroepithelial tissue compartmentalizes, forming retina, ciliary body, and iris. The retina can be further sub-divided into peripheral and central compartments, where the central domain is specialized for higher visual acuity, having a higher ratio and density of cone photoreceptors in most species. RESULTS: Classically, models depict a segregation of the early optic cup into only two domains, neural and non-neural. Recent studies, however, uncovered discrete precursors for central and peripheral retina in the optic vesicle, indicating that the neural retina cannot be considered as a single unit with homogeneous specification and development. Instead, central and peripheral retina may be subject to distinct developmental pathways that underlie their specialization. CONCLUSIONS: This review focuses on lineage relationships in the retina and revisits the historical context for segregation of central and peripheral retina precursors before overt eye morphogenesis.


Subject(s)
Organogenesis/physiology , Retina/embryology , Stem Cells/metabolism , Animals , Humans , Retina/cytology , Stem Cells/cytology
19.
Development ; 140(24): 4959-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24227655

ABSTRACT

The retinal pigment epithelium (RPE) is indispensable for vertebrate eye development and vision. In the classical model of optic vesicle patterning, the surface ectoderm produces fibroblast growth factors (FGFs) that specify the neural retina (NR) distally, whereas TGFß family members released from the proximal mesenchyme are involved in RPE specification. However, we previously proposed that bone morphogenetic proteins (BMPs) released from the surface ectoderm are essential for RPE specification in chick. We now show that the BMP- and Wnt-expressing surface ectoderm is required for RPE specification. We reveal that Wnt signalling from the overlying surface ectoderm is involved in restricting BMP-mediated RPE specification to the dorsal optic vesicle. Wnt2b is expressed in the dorsal surface ectoderm and subsequently in dorsal optic vesicle cells. Activation of Wnt signalling by implanting Wnt3a-soaked beads or inhibiting GSK3ß at optic vesicle stages inhibits NR development and converts the entire optic vesicle into RPE. Surface ectoderm removal at early optic vesicle stages or inhibition of Wnt, but not Wnt/ß-catenin, signalling prevents pigmentation and downregulates the RPE regulatory gene Mitf. Activation of BMP or Wnt signalling can replace the surface ectoderm to rescue MITF expression and optic cup formation. We provide evidence that BMPs and Wnts cooperate via a GSK3ß-dependent but ß-catenin-independent pathway at the level of pSmad to ensure RPE specification in dorsal optic vesicle cells. We propose a new dorsoventral model of optic vesicle patterning, whereby initially surface ectoderm-derived Wnt signalling directs dorsal optic vesicle cells to develop into RPE through a stabilising effect of BMP signalling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Ectoderm/cytology , Retinal Pigment Epithelium/embryology , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Body Patterning , Cell Differentiation , Chick Embryo , Eye/embryology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Microphthalmia-Associated Transcription Factor/biosynthesis , Retinal Pigment Epithelium/metabolism , Smad Proteins/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
20.
Dev Growth Differ ; 55(7): 668-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24024588

ABSTRACT

Elucidating the mechanisms underlying eye development is essential for advancing the medical treatment of eye-related disorders. The primordium of the eye is an optic vesicle (OV), which has a dual potential for generation of the developing neural retina and retinal pigment epithelium. However, the factors that regulate the differentiation of the retinal primordium remain unclear. We have previously shown that overexpression of Lhx1 and Lhx5, members of the LIM-homeobox genes, induced the formation of a second neural retina from the presumptive pigmented retina of the OV. However, the precise timing of Lhx1 expression required for neural retina differentiation has not been clarified. Moreover, RNA interference of Lhx5 has not been previously reported. Here, using a modified electroporation method, we show that, Lhx1 expression in the forebrain around stage 8 is required for neural retina formation. In addition, we have succeeded in the knockdown of Lhx5 expression, resulting in conversion of the neural retina region to a pigment vesicle-like tissue, which indicates that Lhx5 is also required for neural retina differentiation, which correlates temporally with the activity of Lhx1. These results suggest that Lhx1 and Lhx5 in the forebrain regulate neural retina differentiation by suppressing the development of the retinal pigment epithelium, before the formation of the OV.


Subject(s)
Avian Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Prosencephalon/metabolism , Retina/metabolism , Animals , Avian Proteins/genetics , Cell Differentiation/genetics , Chick Embryo , Gene Expression Regulation, Developmental , Immunohistochemistry , LIM-Homeodomain Proteins/genetics , Prosencephalon/embryology , RNA Interference , Retina/cytology , Retina/embryology , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL