Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.490
Filter
1.
Yale J Biol Med ; 97(1): 41-48, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38559457

ABSTRACT

The intricate steps of human ocular embryology are impacted by cellular and genetic signaling pathways and a myriad of external elements that can affect pregnancy, such as environmental, metabolic, hormonal factors, medications, and intrauterine infections. This review focuses on presenting some of these factors to recognize the multifactorial nature of ocular development and highlight their clinical significance. This review is based on English-language articles sourced from PubMed, Web of Science, and Google Scholar; keywords searched included "ocular development in pregnancy," "ocular embryology," "maternal nutrition," "ophthalmic change," and "visual system development." While some animal models show the disruption of ocular embryology from these external factors, there are limited post-birth assessments in human studies. Much remains unknown about the precise mechanisms of how these external factors can disrupt normal ocular development in utero, and more significant research is needed to understand the pathophysiology of these disruptive effects further. Findings in this review emphasize the importance of additional research in understanding the dynamic association between factors impacting gestation and neonatal ocular development, particularly in the setting of limited resources.


Subject(s)
Eye , Maternal Exposure , Animals , Female , Humans , Infant, Newborn , Pregnancy , Eye/embryology
2.
Rev Assoc Med Bras (1992) ; 70(3): e20231167, 2024.
Article in English | MEDLINE | ID: mdl-38656003

ABSTRACT

OBJECTIVE: The aim of this study was to analyze possible alterations (morphological and inflammatory) in the ocular cells of fetuses from mothers with insulin resistance exposed to saturated fatty acids through the period of pregnancy. METHODS: Wistar female rats were induced to develop insulin resistance before pregnancy. Fetuses' skulls were collected on the 20th day of intrauterine life. The rats were separated on the first day of management into two groups according to the diet applied: control group (C): diet containing soybean oil as a source of fat; and saturated fatty acid group (S): diet containing butter as a source of fat. RESULTS: Histological and immunohistochemical analyses have been conducted. The immunohistochemical analyses of interleukin 6, suppressor of cytokine signaling, 3 and signal transducer and activator of transcription 3 did not demonstrate alterations in the expression of proteins in the fetuses of mothers fed with a saturated fatty diet. Moreover, no histopathological changes were noticed between groups. CONCLUSION: The saturated fatty diet does not induce tissue changes or activate the Janus kinase/signal transducer and activator of transcription signaling pathway during eye development in the fetuses of mothers with insulin resistance.


Subject(s)
Insulin Resistance , Janus Kinases , Rats, Wistar , Signal Transduction , Animals , Female , Pregnancy , Signal Transduction/drug effects , Insulin Resistance/physiology , Janus Kinases/metabolism , Fatty Acids/analysis , Dietary Fats/pharmacology , Dietary Fats/adverse effects , Fetus/drug effects , Immunohistochemistry , STAT3 Transcription Factor/metabolism , Interleukin-6/analysis , Interleukin-6/metabolism , Rats , Eye/embryology , Eye/drug effects
3.
J Cell Biol ; 223(2)2024 02 05.
Article in English | MEDLINE | ID: mdl-38126997

ABSTRACT

Lattice cells (LCs) in the developing Drosophila retina change shape before attaining final form. Previously, we showed that repeated contraction and expansion of apical cell contacts affect these dynamics. Here, we describe another factor, the assembly of a Rho1-dependent medioapical actomyosin ring formed by nodes linked by filaments that contract the apical cell area. Cell area contraction alternates with relaxation, generating pulsatile changes in cell area that exert force on neighboring LCs. Moreover, Rho1 signaling is sensitive to mechanical changes, becoming active when tension decreases and cells expand, while the negative regulator RhoGAP71E accumulates when tension increases and cells contract. This results in cycles of cell area contraction and relaxation that are reciprocally synchronized between adjacent LCs. Thus, mechanically sensitive Rho1 signaling controls pulsatile medioapical actomyosin contraction and coordinates cell behavior across the epithelium. Disrupting the kinetics of pulsing can lead to developmental errors, suggesting this process controls cell shape and tissue integrity during epithelial morphogenesis of the retina.


Subject(s)
Actomyosin , Drosophila , Eye , Animals , Actin Cytoskeleton/physiology , Actomyosin/physiology , Cytokinesis , Drosophila/embryology , Morphogenesis , Eye/embryology , rho GTP-Binding Proteins/physiology , Drosophila Proteins/physiology , Retina/cytology
4.
Science ; 378(6621): eabg3679, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36395225

ABSTRACT

The Hippo signaling pathway is widely considered a master regulator of organ growth because of the prominent overgrowth phenotypes caused by experimental manipulation of its activity. Contrary to this model, we show here that removing Hippo transcriptional output did not impair the ability of the mouse liver and Drosophila eyes to grow to their normal size. Moreover, the transcriptional activity of the Hippo pathway effectors Yap/Taz/Yki did not correlate with cell proliferation, and hyperactivation of these effectors induced gene expression programs that did not recapitulate normal development. Concordantly, a functional screen in Drosophila identified several Hippo pathway target genes that were required for ectopic overgrowth but not normal growth. Thus, Hippo signaling does not instruct normal growth, and the Hippo-induced overgrowth phenotypes are caused by the activation of abnormal genetic programs.


Subject(s)
Drosophila melanogaster , Eye , Gene Expression Regulation, Developmental , Hippo Signaling Pathway , Liver , Transcription, Genetic , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Animals , Mice , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye/embryology , Hippo Signaling Pathway/genetics , Liver/embryology , Organ Size , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/metabolism
5.
Cells ; 10(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34572097

ABSTRACT

MT1-MMP/MMP14 belongs to a subgroup of the matrix metalloproteinases family that presents a transmembrane domain, with a cytosolic tail and the catalytic site exposed to the extracellular space. Deficient mice for this enzyme result in early postnatal death and display severe defects in skeletal, muscle and lung development. By using a transgenic line expressing the LacZ reporter under the control of the endogenous Mt1-mmp promoter, we reported a dynamic spatiotemporal expression pattern for Mt1-mmp from early embryonic to perinatal stages during cardiovascular development and brain formation. Thus, Mt1-mmp shows expression in the endocardium of the heart and the truncus arteriosus by E8.5, and is also strongly detected during vascular system development as well as in endothelial cells. In the brain, LacZ reporter expression was detected in the olfactory bulb, the rostral cerebral cortex and the caudal mesencephalic tectum. LacZ-positive cells were observed in neural progenitors of the spinal cord, neural crest cells and the intersomitic region. In the limb, Mt1-mmp expression was restricted to blood vessels, cartilage primordium and muscles. Detection of the enzyme was confirmed by Western blot and immunohistochemical analysis. We suggest novel functions for this metalloproteinase in angiogenesis, endocardial formation and vascularization during organogenesis. Moreover, Mt1-mmp expression revealed that the enzyme may contribute to heart, muscle and brain throughout development.


Subject(s)
Cardiovascular System/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Eye/metabolism , Matrix Metalloproteinase 14/metabolism , Morphogenesis , Nervous System/metabolism , Animals , Cardiovascular System/embryology , Cells, Cultured , Embryo, Mammalian/cytology , Extremities/embryology , Extremities/physiology , Eye/embryology , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Nervous System/embryology
6.
Biochem Soc Trans ; 49(3): 1397-1408, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34196366

ABSTRACT

The Eyes Absent (EYA) transactivator-phosphatase proteins are important contributors to cell-fate determination processes and to the development of multiple organs. The transcriptional regulatory activity as well as the protein tyrosine phosphatase activities of the EYA proteins can independently contribute to proliferation, differentiation, morphogenesis and tissue homeostasis in different contexts. Aberrant EYA levels or activity are associated with numerous syndromic and non-syndromic developmental disorders, as well as cancers. Commensurate with the multiplicity of biochemical activities carried out by the EYA proteins, they impact upon a range of cellular signaling pathways. Here, we provide a broad overview of the roles played by EYA proteins in development, and highlight the molecular signaling pathways known to be linked with EYA-associated organ development and developmental disorders.


Subject(s)
Congenital Abnormalities/genetics , Eye/metabolism , Gene Expression Regulation, Developmental , Kidney/metabolism , Protein Tyrosine Phosphatases/genetics , Trans-Activators/genetics , Animals , Congenital Abnormalities/embryology , Congenital Abnormalities/metabolism , Eye/embryology , Eye/growth & development , Genetic Predisposition to Disease/genetics , Humans , Kidney/embryology , Kidney/growth & development , Mutation , Protein Tyrosine Phosphatases/metabolism , Trans-Activators/metabolism
7.
Dev Biol ; 478: 183-204, 2021 10.
Article in English | MEDLINE | ID: mdl-34216573

ABSTRACT

The mechanisms regulating nervous system development are still unknown for a wide variety of taxa. In insects and vertebrates, bone morphogenetic protein (BMP) signaling plays a key role in establishing the dorsal-ventral (D-V) axis and limiting the neuroectoderm to one side of that axis, leading to speculation about the conserved evolution of centralized nervous systems. Studies outside of insects and vertebrates show a more diverse picture of what, if any role, BMP signaling plays in neural development across Bilateria. This is especially true in the morphologically diverse Spiralia (≈Lophotrochozoa). Despite several studies of D-V axis formation and neural induction in spiralians, there is no consensus for how these two processes are related, or whether BMP signaling may have played an ancestral role in either process. To determine the function of BMP signaling during early development of the spiralian annelid Capitella teleta, we incubated embryos and larvae in BMP4 protein for different amounts of time. Adding exogenous BMP protein to early-cleaving C. teleta embryos had a striking effect on formation of the brain, eyes, foregut, and ventral midline in a time-dependent manner. However, adding BMP did not block brain or VNC formation or majorly disrupt the D-V axis. We identified three key time windows of BMP activity. 1) BMP treatment around birth of the 3rd-quartet micromeres caused the loss of the eyes, radialization of the brain, and a reduction of the foregut, which we interpret as a loss of A- and C-quadrant identities with a possible trans-fate switch to a D-quadrant identity. 2) Treatment after the birth of micromere 4d induced formation of a third ectopic brain lobe, eye, and foregut lobe, which we interpret as a trans-fate switch of B-quadrant micromeres to a C-quadrant identity. 3) Continuous BMP treatment from late cleavage (4d â€‹+ â€‹12 â€‹h) through mid-larval stages resulted in a modest expansion of Ct-chrdl expression in the dorsal ectoderm and a concomitant loss of the ventral midline (neurotroch ciliary band). Loss of the ventral midline was accompanied by a collapse of the bilaterally-symmetric ventral nerve cord, although the total amount of neural tissue was not greatly affected. Our results compared with those from other annelids and molluscs suggest that BMP signaling was not ancestrally involved in delimiting neural tissue to one region of the D-V axis. However, the effects of ectopic BMP on quadrant-identity during cleavage stages may represent a non-axial organizing signal that was present in the last common ancestor of annelids and mollusks. Furthermore, in the last common ancestor of annelids, BMP signaling may have functioned in patterning ectodermal fates along the D-V axis in the trunk. Ultimately, studies on a wider range of spiralian taxa are needed to determine the role of BMP signaling during neural induction and neural patterning in the last common ancestor of this group. Ultimately, these comparisons will give us insight into the evolutionary origins of centralized nervous systems and body plans.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Proteins/metabolism , Polychaeta/embryology , Polychaeta/metabolism , Zebrafish Proteins/pharmacology , Animals , Body Patterning/drug effects , Bone Morphogenetic Proteins/genetics , Brain/embryology , Digestive System/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development , Eye/embryology , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Polychaeta/drug effects , Polychaeta/growth & development , Recombinant Proteins/pharmacology , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism
8.
Biomed Res Int ; 2021: 4982227, 2021.
Article in English | MEDLINE | ID: mdl-34285913

ABSTRACT

PDGFRα signaling is critically important in ocular development. Previous data on PDGFRα lacks an expression map with high spatial and temporal resolution and lineage information. In this study, we aim to present a detailed PDGFRα expression and lineage map from early embryogenesis to adulthood. PDGFRα-CreER; mT/mG reporter mice were analyzed. mEGFP-positive cells contributed to multiple ocular lineages in a spatiotemporally regulated manner. A dynamic PDGFRα expression was identified in corneal stromal cells, lens epithelial cells, lens fiber cells, and retinal astrocytes during the entire period of eye development, while PDGFRα expression in retinal astrocytes from E17.5 onwards and in Müller glial cells was identified within two weeks after birth. By revealing detailed characterization of gene expression and function, we present a comprehensive map of PDGFRα-expressing cells in the eye for a better understanding of PDGFRα signaling's role during eye development.


Subject(s)
Cell Lineage , Eye/cytology , Eye/embryology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Animals, Newborn , Cell Lineage/genetics , Cornea/cytology , Cornea/embryology , Embryo, Mammalian/cytology , Eye/metabolism , Gene Expression Regulation, Developmental , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Retina/cytology
9.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34042968

ABSTRACT

During development, gene expression is tightly controlled to facilitate the generation of the diverse cell types that form the central nervous system. Brahma-related gene 1 (Brg1, also known as Smarca4) is the catalytic subunit of the SWItch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complex that regulates transcription. We investigated the role of Brg1 between embryonic day 6.5 (E6.5) and E14.5 in Sox2-positive neural stem cells (NSCs). Being without major consequences at E6.5 and E14.5, loss of Brg1 between E7.5 and E12.5 resulted in the formation of rosette-like structures in the subventricular zone, as well as morphological alterations and enlargement of neural retina (NR). Additionally, Brg1-deficient cells showed decreased survival in vitro and in vivo. Furthermore, we uncovered distinct changes in gene expression upon Brg1 loss, pointing towards impaired neuron functions, especially those involving synaptic communication and altered composition of the extracellular matrix. Comparison with mice deficient for integrase interactor 1 (Ini1, also known as Smarcb1) revealed that the enlarged NR was Brg1 specific and was not caused by a general dysfunction of the SWI/SNF complex. These results suggest a crucial role for Brg1 in NSCs during brain and eye development.


Subject(s)
Brain/embryology , DNA Helicases/genetics , Eye/embryology , Gene Expression Regulation, Developmental/genetics , Nuclear Proteins/genetics , SMARCB1 Protein/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , DNA Helicases/metabolism , Extracellular Matrix/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Nuclear Proteins/metabolism , Transcription Factors/metabolism
10.
Hum Mol Genet ; 30(17): 1591-1606, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34046667

ABSTRACT

The forkhead transcription factor FOXE3 is critical for vertebrate eye development. Recessive and dominant variants cause human ocular disease but the full range of phenotypes and mechanisms of action for the two classes of variants are unknown. We identified FOXE3 variants in individuals with congenital eye malformations and carried out in vitro functional analysis on selected alleles. Sixteen new recessive and dominant families, including six novel variants, were identified. Analysis of new and previously reported genetic and clinical data demonstrated a broad phenotypic range with an overlap between recessive and dominant disease. Most families with recessive alleles, composed of truncating and forkhead-domain missense variants, had severe corneal opacity (90%; sclerocornea in 47%), aphakia (83%) and microphthalmia (80%), but some had milder features including isolated cataract. The phenotype was most variable for recessive missense variants, suggesting that the functional consequences may be highly dependent on the type of amino acid substitution and its position. When assessed, aniridia or iris hypoplasia were noted in 89% and optic nerve anomalies in 60% of recessive cases, indicating that these defects are also common and may be underrecognized. In dominant pedigrees, caused by extension variants, normal eye size (96%), cataracts (99%) and variable anterior segment anomalies were seen in most, but some individuals had microphthalmia, aphakia or sclerocornea, more typical of recessive disease. Functional studies identified variable effects on the protein stability, DNA binding, nuclear localization and transcriptional activity for recessive FOXE3 variants, whereas dominant alleles showed severe impairment in all areas and dominant-negative characteristics.


Subject(s)
Eye Abnormalities/genetics , Eye/embryology , Forkhead Transcription Factors/genetics , Adolescent , Alleles , Cataract/genetics , Child , Corneal Opacity/genetics , Developmental Disabilities/genetics , Eye/growth & development , Eye Abnormalities/enzymology , Female , Forkhead Transcription Factors/metabolism , Humans , Male , Mutation , Pedigree , Phenotype
11.
EBioMedicine ; 67: 103360, 2021 May.
Article in English | MEDLINE | ID: mdl-33975254

ABSTRACT

Ocular morphogenesis in vertebrates is a highly organized process, orchestrated largely by intrinsic genetic programs that exhibit stringent spatiotemporal control. Alternations in these genetic instructions can lead to hereditary or nonhereditary congenital disorders, a major cause of childhood visual impairment, and contribute to common late-onset blinding diseases. Currently, limited treatment options exist for clinical phenotypes involving eye development. This review summarizes recent advances in our understanding of early-onset ocular disorders and highlights genetic complexities in development and diseases, specifically focusing on coloboma, congenital glaucoma and Leber congenital amaurosis. We also discuss innovative paradigms for potential therapeutic modalities.


Subject(s)
Eye Diseases, Hereditary/genetics , Child , Eye/embryology , Eye/metabolism , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/therapy , Genetic Therapy/methods , Humans , Molecular Targeted Therapy/methods , Stem Cell Transplantation/methods
12.
Dev Biol ; 476: 128-136, 2021 08.
Article in English | MEDLINE | ID: mdl-33811855

ABSTRACT

The basic structure of the eye, which is crucial for visual function, is established during the embryonic process of optic cup morphogenesis. Molecular pathways of specification and patterning are integrated with spatially distinct cell and tissue shape changes to generate the eye, with discrete domains and structural features: retina and retinal pigment epithelium enwrap the lens, and the optic fissure occupies the ventral surface of the eye and optic stalk. Interest in the underlying cell biology of eye morphogenesis has led to a growing body of work, combining molecular genetics and imaging to quantify cellular processes such as adhesion and actomyosin activity. These studies reveal that intrinsic machinery and spatiotemporally specific extrinsic inputs collaborate to control dynamics of cell movements and morphologies. Here we consider recent advances in our understanding of eye morphogenesis, with a focus on the mechanics of eye formation throughout vertebrate systems, including insights and potential opportunities using organoids, which may provide a tractable system to test hypotheses from embryonic models.


Subject(s)
Eye/embryology , Optic Disk/embryology , Actomyosin/metabolism , Animals , Cell Movement , Eye/metabolism , Eye/pathology , Humans , Lens, Crystalline/embryology , Morphogenesis/genetics , Morphogenesis/physiology , Optic Disk/metabolism , Organogenesis/genetics , Organogenesis/physiology , Retina/embryology , Retinal Pigment Epithelium/cytology , Signal Transduction , Vertebrates/physiology
13.
J Vis Exp ; (170)2021 04 05.
Article in English | MEDLINE | ID: mdl-33871454

ABSTRACT

Vertebrate eye development is a complex process that begins near the end of embryo gastrulation and requires the precise coordination of cell migration, proliferation, and differentiation. Time-lapse imagining offers unique insight to the behavior of cells during eye development because it allows us to visualize oculogenesis in vivo. Zebrafish are an excellent model to visualize this process due to their highly conserved vertebrate eye and their ability to develop rapidly and externally while remaining optically transparent. Time-lapse imaging studies of zebrafish eye development are greatly facilitated by use of the transgenic zebrafish line Tg(rx3:GFP). In the developing forebrain, rx3:GFP expression marks the cells of the single eye field, and GFP continues to be expressed as the eye field evaginates to form an optic vesicle, which then invaginates to form an optic cup. High resolution time lapse imaging of rx3:GFP expression, therefore, allows us to track the eye primordium through time as it develops into the retina. Lightsheet microscopy is an ideal method to image ocular morphogenesis over time due to its ability to penetrate thicker samples for fluorescent imaging, minimize photobleaching and phototoxicity, and image at a high speed. Here, a protocol is provided for time-lapse imaging of ocular morphogenesis using a commercially available lightsheet microscope and an image processing workstation to analyze the resulting data. This protocol details the procedures for embryo anesthesia, embedding in low melting temperature agarose, suspension in the imaging chamber, setting up the imaging parameters, and finally analyzing the imaging data using image analysis software. The resulting dataset can provide valuable insights into the process of ocular morphogenesis, as well as perturbations to this process as a result of genetic mutation, exposure to pharmacological agents, or other experimental manipulations.


Subject(s)
Embryonic Development , Eye/embryology , Microscopy/methods , Zebrafish/embryology , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Morphogenesis , Zebrafish/genetics
14.
Gene ; 787: 145622, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33798679

ABSTRACT

We clarified the properties of visual opsin genes in the marbled sole (Pseudopleuronectes yokohamae) by cDNA sequencing, quantification of the opsin gene expression from the larval to the juvenile stage, and measurement of the maximum absorption spectra (λmax) using photopigment reconstitution. In the marbled sole eye, at least eight visual opsin genes, lws, rh2-a, rh2-b, rh2-c, sws2a, sws2b, sws1, and rh1, were expressed. Quantitative RT-PCR analysis revealed that the expression of opsin genes increased (lws, rh2-c, sws2a, and rh1) or decreased (rh2-a, rh2-b, sws2b, and sws1) from the larval to the juvenile stage. Notably, rh2-a expression was observed only in pre- to mid-metamorphic stage larvae and disappeared after metamorphosis. Thus, pre-metamorphism-specific expression of rh2-a in the marbled sole suggests that its function is restricted to the developmental stage. The reconstituted RH2-A opsin λmax was 470 nm, which is typical of acanthopterygian species. These results strongly suggest that mid-wavelength-sensitive rh2-a expression was diminished drastically in the marbled sole, probably resulting in a shift of spectral sensitivity during its metamorphosis from the larval to the juvenile stage.


Subject(s)
Flounder/genetics , Rhodopsin/genetics , Animals , Eye/embryology , Eye/metabolism , Flounder/embryology , Larva/genetics , Larva/growth & development , Phylogeny , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Spectrophotometry , Transcriptome
15.
PLoS Genet ; 17(3): e1009458, 2021 03.
Article in English | MEDLINE | ID: mdl-33755662

ABSTRACT

Precise regulation of ocular size is a critical determinant of normal visual acuity. Although it is generally accepted that ocular growth relies on a cascade of signaling events transmitted from the retina to the sclera, the factors and mechanism(s) involved are poorly understood. Recent studies have highlighted the importance of the retinal secreted serine protease PRSS56 and transmembrane glycoprotein MFRP, a factor predominantly expressed in the retinal pigment epithelium (RPE), in ocular size determination. Mutations in PRSS56 and MFRP constitute a major cause of nanophthalmos, a condition characterized by severe reduction in ocular axial length/extreme hyperopia. Interestingly, common variants of these genes have been implicated in myopia, a condition associated with ocular elongation. Consistent with these findings, mice with loss of function mutation in PRSS56 or MFRP exhibit a reduction in ocular axial length. However, the molecular network and cellular processes involved in PRSS56- and MFRP-mediated ocular axial growth remain elusive. Here, we show that Adamts19 expression is significantly upregulated in the retina of mice lacking either Prss56 or Mfrp. Importantly, using genetic mouse models, we demonstrate that while ADAMTS19 is not required for ocular growth during normal development, its inactivation exacerbates ocular axial length reduction in Prss56 and Mfrp mutant mice. These results suggest that the upregulation of retinal Adamts19 is part of an adaptive molecular response to counteract impaired ocular growth. Using a complementary genetic approach, we show that loss of PRSS56 or MFRP function prevents excessive ocular axial growth in a mouse model of early-onset myopia caused by a null mutation in Irbp, thus, demonstrating that PRSS56 and MFRP are also required for pathological ocular elongation. Collectively, our findings provide new insights into the molecular network involved in ocular axial growth and support a role for molecular crosstalk between the retina and RPE involved in refractive development.


Subject(s)
ADAMTS Proteins/genetics , Eye Proteins/genetics , Eye/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Membrane Proteins/genetics , Organogenesis/genetics , Serine Proteases/genetics , ADAMTS Proteins/metabolism , Animals , Biomarkers , Eye/embryology , Eye/growth & development , Eye Proteins/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Retinol-Binding Proteins/genetics , Serine Proteases/metabolism , Signal Transduction
16.
Nat Commun ; 12(1): 1447, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664263

ABSTRACT

Identifying the genetic factors that underlie complex traits is central to understanding the mechanistic underpinnings of evolution. Cave-dwelling Astyanax mexicanus populations are well adapted to subterranean life and many populations appear to have evolved troglomorphic traits independently, while the surface-dwelling populations can be used as a proxy for the ancestral form. Here we present a high-resolution, chromosome-level surface fish genome, enabling the first genome-wide comparison between surface fish and cavefish populations. Using this resource, we performed quantitative trait locus (QTL) mapping analyses and found new candidate genes for eye loss such as dusp26. We used CRISPR gene editing in A. mexicanus to confirm the essential role of a gene within an eye size QTL, rx3, in eye formation. We also generated the first genome-wide evaluation of deletion variability across cavefish populations to gain insight into this potential source of cave adaptation. The surface fish genome reference now provides a more complete resource for comparative, functional and genetic studies of drastic trait differences within a species.


Subject(s)
Adaptation, Physiological/genetics , Characidae/embryology , Characidae/genetics , Eye/embryology , Multifactorial Inheritance/genetics , Animals , Biological Evolution , Caves , Chromosome Mapping , Evolution, Molecular , Gene Editing , Genome/genetics , Homeodomain Proteins/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Quantitative Trait Loci/genetics
17.
Dev Biol ; 476: 41-52, 2021 08.
Article in English | MEDLINE | ID: mdl-33745943

ABSTRACT

Understanding molecular mechanisms of sexually dimorphic organ growth is a fundamental problem of developmental biology. Recent quantitative studies showed that the Drosophila compound eye is a convenient model to study the determination of the final organ size. In Drosophila, females have larger eyes than males and this is evident even after correction for the larger body size. Moreover, female eyes include more ommatidia (photosensitive units) than male eyes and this difference is specified at the third larval instar in the eye primordia called eye imaginal discs. This may result in different visual capabilities between the two sexes and have behavioral consequences. Despite growing evidence on the genetic bases of eye size variation between different Drosophila species and strains, mechanisms responsible for within-species sexual dimorphism still remain elusive. Here, we discuss a presumptive crosstalk between the sex determination cascade and major signaling pathways during dimorphic eye development. Male- and female-specific isoforms of Doublesex (Dsx) protein are known to control sex-specific differentiation in the somatic tissues. However, no data on Dsx function during eye disc growth and patterning are currently available. Remarkably, Sex lethal (Sxl), the sex determination switch protein, was shown to directly affect Hedgehog (Hh) and Notch (N) signaling in the Drosophila wing disc. The similarity of signaling pathways involved in the wing and eye disc growth suggests that Sxl might be integrated into regulation of eye development. Dsx role in the eye disc requires further investigation. We discuss currently available data on sex-biased gene expression in the Drosophila eye and highlight perspectives for future studies.


Subject(s)
Eye/embryology , Sex Determination Processes/genetics , Sex Differentiation/genetics , Animals , DNA-Binding Proteins/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development/genetics , Eye/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/genetics , Male , RNA-Binding Proteins/genetics , Sex Characteristics , Sex Determination Processes/physiology , Sex Factors , Signal Transduction/genetics , Signal Transduction/physiology
18.
Biochem Soc Trans ; 49(1): 327-340, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33616626

ABSTRACT

Developmental regulation of the vertebrate visual system has been a focus of investigation for generations as understanding this critical time period has direct implications on our understanding of congenital blinding disease. The majority of studies to date have focused on transcriptional regulation mediated by morphogen gradients and signaling pathways. However, recent studies of post translational regulation during ocular development have shed light on the role of the ubiquitin proteasome system (UPS). This rather ubiquitous yet highly diverse system is well known for regulating protein function and localization as well as stability via targeting for degradation by the 26S proteasome. Work from many model organisms has recently identified UPS activity during various milestones of ocular development including retinal morphogenesis, retinal ganglion cell function as well as photoreceptor homeostasis. In particular work from flies and zebrafish has highlighted the role of the E3 ligase enzyme family, Seven in Absentia Homologue (Siah) during these events. In this review, we summarize the current understanding of UPS activity during Drosophila and vertebrate ocular development, with a major focus on recent findings correlating Siah E3 ligase activity with two major developmental stages of vertebrate ocular development, retinal morphogenesis and photoreceptor specification and survival.


Subject(s)
Eye/embryology , Nuclear Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Drosophila , Eye/growth & development , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology , Vertebrates , Zebrafish
19.
Nat Commun ; 12(1): 1125, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602923

ABSTRACT

Conditional gene inactivation is a powerful tool to determine gene function when constitutive mutations result in detrimental effects. The most commonly used technique to achieve conditional gene inactivation employs the Cre/loxP system and its ability to delete DNA sequences flanked by two loxP sites. However, targeting a gene with two loxP sites is time and labor consuming. Here, we show Cre-Controlled CRISPR (3C) mutagenesis to circumvent these issues. 3C relies on gRNA and Cre-dependent Cas9-GFP expression from the same transgene. Exogenous or transgenic supply of Cre results in Cas9-GFP expression and subsequent mutagenesis of the gene of interest. The recombined cells become fluorescently visible enabling their isolation and subjection to various omics techniques. Hence, 3C mutagenesis provides a valuable alternative to the production of loxP-flanked alleles. It might even enable the conditional inactivation of multiple genes simultaneously and should be applicable to other model organisms amenable to single integration transgenesis.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Silencing , Integrases/metabolism , Mutagenesis/genetics , Zebrafish/genetics , Animals , Base Sequence , Eye/embryology , Eye/metabolism , Green Fluorescent Proteins/metabolism , Monophenol Monooxygenase/genetics , Pigmentation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transgenes
20.
Dev Dyn ; 250(8): 1056-1073, 2021 08.
Article in English | MEDLINE | ID: mdl-33570754

ABSTRACT

BACKGROUND: The male-abnormal 21 like (MAB21L) genes are important in human ocular development. Homozygous loss of MAB21L1 leads to corneal dystrophy in all affected individuals along with cataracts and buphthalmos in some. The molecular function and downstream pathways of MAB21L factors are largely undefined. RESULTS: We generated the first mab21l1 zebrafish mutant carrying a putative loss-of-function allele, c.107delA p.(Lys36Argfs*7). At the final stages of embryonic development, homozygous mab21l1c.107delA fish displayed enlarged anterior chambers and corneal thinning which progressed with age. Additional studies revealed increased cell death in the mutant corneas, transformation of the cornea into a skin-like epithelium, and progressive lens degeneration with development of fibrous masses in the anterior chamber. RNA-seq of wild-type and mutant ocular transcriptomes revealed significant changes in expression of several genes, including irf1a and b, stat1, elf3, krt17, tlr9, and loxa associated with immunity and/or corneal function. Abnormal expression of lysyl oxidases have been previously linked with corneal thinning, fibrosis, and lens defects in mammals, suggesting a role for loxa misexpression in the progressive mab21l1c.107delA eye phenotype. CONCLUSIONS: Zebrafish mab21l1 is essential for normal corneal development, similar to human MAB21L1. The identified molecular changes in mab21l1c.107delA mutants provide the first clues about possible affected pathways.


Subject(s)
Eye/embryology , Homeodomain Proteins/genetics , Organogenesis/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Cornea/embryology , Cornea/metabolism , Embryonic Development/genetics , Eye/metabolism , Homeodomain Proteins/metabolism , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Phenotype , Zebrafish , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...