Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
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
2.
J Neurosci ; 39(13): 2398-2415, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30692221

ABSTRACT

Primary cilia are essential for CNS development. In the mouse, they play a critical role in patterning the spinal cord and telencephalon via the regulation of Hedgehog/Gli signaling. However, despite the frequent disruption of this signaling pathway in human forebrain malformations, the role of primary cilia in forebrain morphogenesis has been little investigated outside the telencephalon. Here we studied development of the diencephalon, hypothalamus and eyes in mutant mice in which the Ftm/Rpgrip1l ciliopathy gene is disrupted. At the end of gestation, Ftm-/- fetuses displayed anophthalmia, a reduction of the ventral hypothalamus and a disorganization of diencephalic nuclei and axonal tracts. In Ftm-/- embryos, we found that the ventral forebrain structures and the rostral thalamus were missing. Optic vesicles formed but lacked the optic cups. In Ftm-/- embryos, Sonic hedgehog (Shh) expression was virtually lost in the ventral forebrain but maintained in the zona limitans intrathalamica (ZLI), the mid-diencephalic organizer. Gli activity was severely downregulated but not lost in the ventral forebrain and in regions adjacent to the Shh-expressing ZLI. Reintroduction of the repressor form of Gli3 into the Ftm-/- background restored optic cup formation. Our data thus uncover a complex role of cilia in development of the diencephalon, hypothalamus and eyes via the region-specific control of the ratio of activator and repressor forms of the Gli transcription factors. They call for a closer examination of forebrain defects in severe ciliopathies and for a search for ciliopathy genes as modifiers in other human conditions with forebrain defects.SIGNIFICANCE STATEMENT The Hedgehog (Hh) signaling pathway is essential for proper forebrain development as illustrated by a human condition called holoprosencephaly. The Hh pathway relies on primary cilia, cellular organelles that receive and transduce extracellular signals and whose dysfunctions lead to rare inherited diseases called ciliopathies. To date, the role of cilia in the forebrain has been poorly studied outside the telencephalon. In this paper we study the role of the Ftm/Rpgrip1l ciliopathy gene in mouse forebrain development. We uncover complex functions of primary cilia in forebrain morphogenesis through region-specific modulation of the Hh pathway. Our data call for further examination of forebrain defects in ciliopathies and for a search for ciliopathy genes as modifiers in human conditions affecting forebrain development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hedgehog Proteins/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Zinc Finger Protein Gli3/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Eye/embryology , Eye/metabolism , Hypothalamus/embryology , Hypothalamus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Thalamus/embryology , Thalamus/metabolism
3.
Annu Rev Vis Sci ; 4: 263-285, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29856937

ABSTRACT

The thalamocortical pathway is the main route of communication between the eye and the cerebral cortex. During embryonic development, thalamocortical afferents travel to L4 and are sorted by receptive field position, eye of origin, and contrast polarity (i.e., preference for light or dark stimuli). In primates and carnivores, this sorting involves numerous afferents, most of which sample a limited region of the binocular field. Devoting abundant thalamocortical resources to process a limited visual field has a clear advantage: It allows many stimulus combinations to be sampled at each spatial location. Moreover, the sampling efficiency can be further enhanced by organizing the afferents in a cortical grid for eye input and contrast polarity. We argue that thalamocortical interactions within this eye-polarity grid can be used to represent multiple stimulus combinations found in nature and to build an accurate cortical map for multidimensional stimulus space.


Subject(s)
Neural Pathways/physiology , Retinal Neurons/physiology , Thalamus/physiology , Visual Cortex/physiology , Visual Perception/physiology , Brain Mapping , Eye/embryology , Humans , Neural Pathways/embryology , Neurons, Afferent/physiology , Thalamus/embryology , Visual Cortex/embryology , Visual Fields/physiology , Visual Pathways/physiology
4.
Int J Dev Biol ; 61(3-4-5): 149-157, 2017.
Article in English | MEDLINE | ID: mdl-28621412

ABSTRACT

The zebrafish has become a model of choice in fundamental and applied life sciences and is widely used in various fields of biomedical research as a human disease model for cancer, metabolic and neurodegenerative diseases, and regenerative medicine. The transparency of the zebrafish embryo allows real-time visualization of the development and morphogenesis of practically all of its tissues and organs. Zebrafish are amenable to genetic manipulation, for which innovative genetic and molecular techniques are constantly being introduced. These include the study of gene function and regulation using gene knockdown, knockout and knock-in, as well as transgenesis and tissue-specific genetic perturbations. Complementing this genetic toolbox, the zebrafish exhibits measurable behavioral and hormonal responses already at the larval stages, providing a viable vertebrate animal model for high-throughput drug screening and chemical genetics. With the available tools of the genomic era and the abundance of disease-associated human genes yet to be explored, the zebrafish model is becoming the preferred choice in many studies. Its advantages and potential are being increasingly recognized within the Israeli scientific community, and its use as a model system for basic and applied science has expanded in Israel in recent years. Since the first zebrafish-focused laboratory was introduced at Tel Aviv University 16 years ago, seven more zebrafish-centric research groups have been established, along with more than two dozen academic research groups and three bio-medical companies that are now utilizing this model.


Subject(s)
Biomedical Research/trends , Developmental Biology/trends , Zebrafish/embryology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Aquaculture , Behavior, Animal , Brain/embryology , Cell Cycle , Cell Division , Circadian Rhythm , Developmental Biology/history , Disease Models, Animal , Erythropoiesis , Eye/embryology , Genomics , History, 20th Century , History, 21st Century , Humans , Hypothalamus/metabolism , Inflammation , Israel , Lipids/chemistry , Microglia , Microscopy, Fluorescence , Neoplasms , Neurosecretory Systems/embryology , Phenotype , Reproducibility of Results , Sleep
5.
Proc Natl Acad Sci U S A ; 113(51): 14710-14715, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27911769

ABSTRACT

The retinal pigment epithelium (RPE) is a monolayer of pigmented cells that requires an active metabolism to maintain outer retinal homeostasis and compensate for oxidative stress. Using 13C metabolic flux analysis in human RPE cells, we found that RPE has an exceptionally high capacity for reductive carboxylation, a metabolic pathway that has recently garnered significant interest because of its role in cancer cell survival. The capacity for reductive carboxylation in RPE exceeds that of all other cells tested, including retina, neural tissue, glial cells, and a cancer cell line. Loss of reductive carboxylation disrupts redox balance and increases RPE sensitivity to oxidative damage, suggesting that deficiencies of reductive carboxylation may contribute to RPE cell death. Supporting reductive carboxylation by supplementation with an NAD+ precursor or its substrate α-ketoglutarate or treatment with a poly(ADP ribose) polymerase inhibitor protects reductive carboxylation and RPE viability from excessive oxidative stress. The ability of these treatments to rescue RPE could be the basis for an effective strategy to treat blinding diseases caused by RPE dysfunction.


Subject(s)
Carbon/chemistry , Eye/embryology , Ketoglutaric Acids/chemistry , Macular Degeneration/metabolism , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Aged, 80 and over , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival , Fatty Acids/chemistry , Female , HeLa Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Isocitrate Dehydrogenase/metabolism , Macular Degeneration/pathology , Mice , NAD/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress , Oxygen/chemistry , Poly(ADP-ribose) Polymerases/metabolism
6.
FASEB J ; 28(4): 1854-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24421398

ABSTRACT

Patients with congenital disorder of glycosylation (CDG), type Ib (MPI-CDG or CDG-Ib) have mutations in phosphomannose isomerase (MPI) that impair glycosylation and lead to stunted growth, liver dysfunction, coagulopathy, hypoglycemia, and intestinal abnormalities. Mannose supplements correct hypoglycosylation and most symptoms by providing mannose-6-P (Man-6-P) via hexokinase. We generated viable Mpi hypomorphic mice with residual enzymatic activity comparable to that of patients, but surprisingly, these mice appeared completely normal except for modest (~15%) embryonic lethality. To overcome this lethality, pregnant dams were provided 1-2% mannose in their drinking water. However, mannose further reduced litter size and survival to weaning by 40 and 66%, respectively. Moreover, ~50% of survivors developed eye defects beginning around midgestation. Mannose started at birth also led to eye defects but had no effect when started after eye development was complete. Man-6-P and related metabolites accumulated in the affected adult eye and in developing embryos and placentas. Our results demonstrate that disturbing mannose metabolic flux in mice, especially during embryonic development, induces a highly specific, unanticipated pathological state. It is unknown whether mannose is harmful to human fetuses during gestation; however, mothers who are at risk for having MPI-CDG children and who consume mannose during pregnancy hoping to benefit an affected fetus in utero should be cautious.


Subject(s)
Blindness/etiology , Dietary Supplements/toxicity , Mannose-6-Phosphate Isomerase/metabolism , Mannose/toxicity , Animals , Blindness/genetics , Blindness/metabolism , Blotting, Western , Cells, Cultured , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Eye/embryology , Eye/growth & development , Eye/metabolism , Female , Humans , Immunohistochemistry , Male , Mannose/blood , Mannose/metabolism , Mannose-6-Phosphate Isomerase/genetics , Mannosephosphates/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Placenta/drug effects , Placenta/embryology , Placenta/metabolism , Pregnancy
7.
Mol Vis ; 17: 1473-84, 2011.
Article in English | MEDLINE | ID: mdl-21677791

ABSTRACT

PURPOSE: We recently demonstrated that molecular therapy using aminoglycosides can overcome the underlying genetic defect in two zebrafish models of ocular coloboma and showed abnormal cell death to be a key feature associated with the optic fissure closure defects. In further studies to identify molecular therapies for this common congenital malformation, we now examine the effects of anti-apoptotic compounds in zebrafish models of ocular coloboma in vivo. METHODS: Two ocular coloboma zebrafish lines (pax2.1/noi(tu29a) and lamb1/gup(m189)) were exposed to diferuloylmethane (curcumin) or benzyloxycarbonyl-Val-Ala-Asp(Ome)-fluoromethylketone (zVAD-fmk; a pan-caspase inhibitor) for up to 8 days post-fertilization. The effects of these compounds were assessed by morphology, histology, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and western blot analysis. RESULTS: The size of the coloboma in gup zebrafish mutants treated with diferuloylmethane was greatly reduced. In treated mutants a reduction in TUNEL staining and a 67% decrease in activated caspase-3 protein were observed. The release of cytochrome c from the mitochondria into the cytosol was reduced fourfold by in vivo diferuloylmethane treatment, suggesting that the drug was acting to inhibit the intrinsic apoptotic pathway. Inhibition of caspases directly with zVAD-fmk also resulted in a similar reduction in coloboma phenotype. Treatment with either diferuloylmethane or zVAD-fmk resulted in a statistically significant 1.4 fold increase in length of survival of these mutant zebrafish (p<0.001), which normally succumb to the lethal genetic mutation. In contrast, the coloboma phenotype in noi zebrafish mutants did not respond to either diferuloylmethane or zVAD-fmk exposure, even though inhibition of apoptotic cell death was observed by a reduction in TUNEL staining. CONCLUSIONS: The differential sensitivity to anti-apoptotic agents in lamb1-deficient and pax2.1-deficient zebrafish models, suggests that apoptotic cell death is not a final common pathway in all ocular coloboma genotypes. When considering anti-cell death therapies for ocular colobomatous defects attention should be paid to the genotype under investigation.


Subject(s)
Cell Death/genetics , Coloboma , Curcumin/pharmacology , Eye/metabolism , Zebrafish/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/drug effects , Coloboma/embryology , Coloboma/genetics , Coloboma/metabolism , Coloboma/pathology , Cytochromes c/analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Nonmammalian , Eye/embryology , Eye/pathology , Genetic Variation , In Situ Nick-End Labeling , Longevity/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mutation , Phenotype , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
Int J Dev Biol ; 53(4): 469-82, 2009.
Article in English | MEDLINE | ID: mdl-19378250

ABSTRACT

Pax transcription factors are evolutionarily conserved regulators of eye development and can be distinguished on the basis of three functional domains: two DNA-binding domains (the paired domain and the paired-type homeodomain), and the octapeptide motif. PaxB of the eyed cubozoan jellyfish, Tripedalia cystophora, is characterized by a Pax2-like paired domain and octapeptide, and a Pax6-like homeodomain. In mice, functionally distinct Pax6 and Pax2 proteins have unique as well as redundant roles in eye morphogenesis. Here, we show that expression of the jellyfish PaxB gene in mouse embryonic eye tissues impairs normal development of lens and retina. Our data show that PaxB misexpression leads to a downregulation of endogenous Pax6 protein in the prospective lens and in subsets of cells within the inner nuclear layer of transgenic retina. In addition to Pax6 downregulation, the expression of PaxB leads to an almost complete loss of amacrine cells in the adult transgenic retina, a phenotype that differs from a loss-of-function of the Pax6 gene. The present data suggest that PaxB, due to its Pax2-like paired domain and Pax-6 like homeodomain, disturbs the transcriptional network regulated by Pax6 in the developing lens and retina. Taken together, our data suggest that molecular properties of individual mouse Pax2 and Pax6 proteins are essential determinants of mouse eye development and cannot be substituted for by jellyfish PaxB which possesses elements of vertebrate Pax2 and Pax6.


Subject(s)
Eye Proteins/metabolism , Eye/metabolism , Homeodomain Proteins/metabolism , Otx Transcription Factors/metabolism , PAX2 Transcription Factor/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Scyphozoa/metabolism , Animals , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Eye/embryology , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Otx Transcription Factors/genetics , PAX2 Transcription Factor/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Phenotype , Repressor Proteins/genetics , Scyphozoa/genetics
9.
J Toxicol Environ Health A ; 70(18): 1542-55, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17710614

ABSTRACT

Exposure of the early life stages of fish to oil sands constituents is associated with mortality and larval malformations such as edemas, hemorrhages, and skeletal, craniofacial, and eye defects. In fathead minnow (Pimephales promelas) and white sucker (Catostomus commersoni) larvae, indices of total eye pathology increased significantly following oil sands exposure. Structural, cytoplasmic, inflammatory, and degenerative eye alterations included poor retinal differentiation, microphthalmia, optic fissures, dysphasic retinas and lenses, inflammatory infiltrates, retinal epithelial lifting, and necrotic foci. Cytochrome P-4501A (CYP1A) was expressed in ocular (retina, lens) and kidney endothelial tissues, as indicated by immunohistochemistry. Although the kinetics of exposure-response curves for mortality and CYP1A expression were similar in both species, species differences in the magnitude and sensitivity of the responses were observed. Oil sands were twofold more toxic to fathead minnows (TPAH LC50 = 47-330 microg/g) than to white sucker (TPAH LC50 = 95-860 microg/g) larvae. For both species, larval mortality was significantly related to CYP1A protein concentrations in kidneys, and severity of these effects rose with oil sands exposure. The relationships among eye damage, mortality, and CYP1A indices warrants further investigation, and may lead to the use of CYP1A induction as an indicator of adverse effects rather than just contaminant exposure.


Subject(s)
Cyprinidae/embryology , Cytochrome P-450 CYP1A1/metabolism , Eye/drug effects , Hydrocarbons/toxicity , Industrial Waste/adverse effects , Petroleum , Animals , Cyprinidae/metabolism , Eye/embryology , Eye/enzymology , Eye/pathology , Geologic Sediments , Kidney/drug effects , Kidney/enzymology , Larva/drug effects , Larva/enzymology , Larva/growth & development , Lethal Dose 50 , Polycyclic Aromatic Hydrocarbons/analysis , Polycyclic Aromatic Hydrocarbons/toxicity , Silicon Dioxide , Waste Disposal, Fluid
10.
Hum Reprod ; 22(1): 280-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16959805

ABSTRACT

BACKGROUND: Although catechins are known to be powerful antioxidants, no reports have shown their transport to fetal organs. We investigated the distribution of catechins in fetal rat organs after maternal exposure to green tea extract (GTE). METHODS: GTE (550 mg/kg) or water was fed orally to pregnant dams at 15.5 days of gestation, the dams were sacrificed and fetal organs were dissected 0, 0.5, 1, 2, 3, 5, and 8 h later. Catechins and catechin gallates were determined by high-performance liquid chromatography (HPLC) after solid-phase extraction. RESULTS: In the GTE-treated group, catechins were detected in most of the fetal organs studied, including the brain, eyes, heart, lungs, kidneys and liver but not in the control group. The first peak times (T(max)) were about 0.5-1 h. The maximum concentrations (C(max)) of catechins in the fetal eye were about 2-10 times higher than in the other organs, ranging from 249 pmol/g for epicatechin (EC) to 831 pmol/g for epigallocatechin gallate (EGCG). Catechin gallates were generally more readily taken up by fetal organs than catechins. EGCG had the highest level of uptake according to area under the curve (AUC) plots and the highest C(max) in all organs. CONCLUSIONS: Various fetal organs had low but significant levels of catechins after GTE intake by the dams, and organ levels were found to be related to catechin structure. EGCG could be a potential candidate for antioxidant supplementation of the fetus in utero.


Subject(s)
Catechin/metabolism , Fetus/metabolism , Maternal-Fetal Exchange , Animals , Brain/embryology , Brain/metabolism , Camellia sinensis/chemistry , Catechin/analogs & derivatives , Catechin/blood , Eye/embryology , Eye/metabolism , Female , Fetal Heart/metabolism , Kidney/embryology , Kidney/metabolism , Liver/embryology , Liver/metabolism , Lung/embryology , Lung/metabolism , Plant Extracts/metabolism , Pregnancy , Rats , Tissue Distribution
11.
Fly (Austin) ; 1(4): 235-7, 2007.
Article in English | MEDLINE | ID: mdl-18820442

ABSTRACT

Apical basal cell polarity is a fundamental feature of all epithelial cells. Identification of the genes involved in the polarization of epithelial cells has begun to reveal the mechanisms underlying the establishment and maintenance of cell polarity. An important issue is to understand the molecular basis for localization of cell polarity proteins in the context of the developing organism. Bazooka (Baz, Drosophila homolog of Par-3) plays a crucial role in organizing cell polarity in several different tissues. In the ovarian follicle epithelium, Par-1 protein kinase regulates Baz localization to the apical cell cortex by excluding phosphorylated Baz from the lateral region. In photoreceptor cells of retinal epithelium, Baz is targeted to the adherens junction (AJ) instead of the apical domain. Our study suggests that in photoreceptors, Par-1 blocks the localization of Baz to AJ whereas protein phosphatase 2A (PP2A) promotes Baz localization by antagonizing the Par-1 effects. In this extra view, we provide a brief overview and perspective of our findings on the antagonistic function of Par-1 and PP2A in Baz localization during photoreceptor morphogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Photoreceptor Cells, Invertebrate/embryology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Polarity , Drosophila/metabolism , Drosophila Proteins/analysis , Eye/embryology , Glycogen Synthase Kinase 3 , Intracellular Signaling Peptides and Proteins/analysis , Phosphorylation
12.
Dev Growth Differ ; 48(9): 575-85, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17118012

ABSTRACT

Using a subtracted Xenopus cDNA library based on the differential sensitivity of anterior and posterior genes to retinoic acid, we isolated a novel Xenopus nuclear GTP-binding protein (XGB). XGB is expressed prominently in the optic primordia at the tailbud stage. The N-terminal region of XGB contains a set of GTP-binding protein motifs, and the C-terminal region contains two putative nuclear localization signals and two coiled regions. A GFP-XGB fusion protein was expressed in the nucleus of NIH3T3 cells where it bound to subnuclear structures. Truncated C-terminal constructs of XGB containing both nuclear localization signal(s) and coiled region(s) suppressed eye formation, whereas neither the N-terminal construct nor constructs with a mutated GTP-binding protein motif affected eye formation. Expression of Pax6 and Rx1 genes, which are crucial for eye development, was reduced in embryos overexpressing the C-terminal constructs of XGB. Suppression of Pax6 and Rx1 at earlier developmental stages as well as perturbation of eye formation at later stages was counteracted by co-expression of wild-type XGB. We conclude that XGB plays a role in the formation of optic primordia through activation of at least two eye field transcription factors.


Subject(s)
Eye/embryology , GTP-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Amino Acid Sequence , Animals , DNA, Complementary , Embryo, Nonmammalian , Embryonic Development/genetics , Eye/metabolism , GTP-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Molecular Sequence Data , Xenopus/genetics , Xenopus Proteins/genetics
13.
Mol Vis ; 12: 1250-8, 2006 Oct 26.
Article in English | MEDLINE | ID: mdl-17110908

ABSTRACT

PURPOSE: To identify the changes in zebrafish embryonic ocular development after early growth response factor 1 (Egr1) gene knockdown by Egr1-specific translation inhibitor, morpholino oligonucleotides (MO). METHODS: Two kinds of Egr1-MO were microinjected separately with various dosages into one to four celled zebrafish embryos to find an optimal dose generating an acceptable mortality rate and high frequency of specific phenotype. Chordin-MO served as the positive control; a 5 mismatch MO of Egr1-MO1 and a nonspecific MO served as negative controls. We graded the Egr1 morphants according to their gross abnormalities, and measured their ocular dimensions accordingly. Western blot analysis and synthetic Egr1 mRNA rescue experiments confirmed whether the deformities were caused by Egr1 gene knockdown. Histological examination and three kinds of immunohistochemical staining were applied to identify glutamate receptor one expression in retinal ganglion cells and amacrine cells, to recognize acetylated alpha-tubulin expression which indicated axonogenesis, and to label photoreceptor cells with zpr-1 antibody. RESULTS: After microinjection of 8 ng Egr1-MO1 or 2 ng Egr1-MO2, 81.8% and 97.3% of larvae at 72 h postfertilization had specific defects, respectively. The gross phenotype included string-like heart, flat head, and deformed tail. The more severely deformed larvae had smaller eyes and pupils. Co-injection of 8 ng Egr1-MO1 and supplementary 12 pg synthetic Egr1 mRNA reduced the gross abnormality rate from 84.4% to 29.7%, and decreased the severity of deformities. Egr1 protein appeared in the wildtype and rescued morphants, but was lacking in the Egr1 morphants with specific deformities. Lenses of Egr1 morphants were smaller and had some residual nucleated lens fiber cells. Morphants' retinal cells arranged disorderly and compactly with thin plexiform layers. Immunohistochemical studies showed that morphants had a markedly decreased number of mature retinal ganglion cells, amacrine cells, and photoreceptor cells. Retinal axonogenesis was prominently reduced in morphants. CONCLUSIONS: The Egr1 gene plays an important role in zebrafish embryonic oculogenesis. Ocular structures including lens and retina were primitive and lacked appropriate differentiation. Such arrested retinal and lenticular development in Egr1 morphants resulted in microphthalmos.


Subject(s)
Early Growth Response Protein 1/genetics , Eye/embryology , Gene Deletion , Zebrafish/embryology , Animals , Embryo, Nonmammalian , Embryonic Development/drug effects , Eye/pathology , Eye Abnormalities/embryology , Eye Abnormalities/pathology , Eye Abnormalities/prevention & control , Gene Expression , Immunohistochemistry/methods , Lens, Crystalline/embryology , Lens, Crystalline/pathology , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/pharmacology , Retina/embryology , Retina/pathology , Staining and Labeling
14.
Dev Dyn ; 235(3): 594-605, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16342201

ABSTRACT

The Sonic hedgehog (Shh) signaling pathway plays a key role in the development of the vertebrate central nervous system, including the eye. This pathway is mediated by the Gli transcription factors (Gli1, Gli2, and Gli3) that differentially activate and repress the expression of specific downstream target genes. In this study, we investigated the roles of the three vertebrate Glis in mediating midline Shh signaling in early ocular development. We examined the ocular phenotypes of Shh and Gli combination mutant mouse embryos and monitored proximodistal and dorsoventral patterning by the expression of specific eye development regulatory genes using in situ hybridization. We show that midline Shh signaling relieves the repressor activity of Gli3 adjacent to the midline and then promotes eye pattern formation through the nonredundant activities of all three Gli proteins. Gli3, in particular, is required to specify the dorsal optic stalk and to define the boundary between the optic stalk and the optic cup.


Subject(s)
Body Patterning/genetics , Eye/embryology , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism , Animals , Eye/anatomy & histology , Gene Expression Regulation, Developmental , Hedgehog Proteins , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Neurologic Mutants , Mutation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Optic Nerve/anatomy & histology , Optic Nerve/embryology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/physiology , Trans-Activators/genetics , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
15.
Dev Biol ; 270(2): 336-49, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15183718

ABSTRACT

Two alleles of an eyeless mutant, chokh (chk), were identified in ongoing zebrafish F(3) mutagenesis screens. Morphologically, chk mutants can be identified at 15 h post-fertilization by the failure of optic primordia to evaginate from the forebrain. The chk phenotype appears specific, as marker genes in the forebrain, midbrain, and pineal are expressed in normal temporal, spatial, and circadian patterns. Sequence analysis of the chk alleles revealed nonsense or missense mutations in the rx3 homeobox. Rx genes encode paired-type homeodomain transcription factors known to be key regulators of eye development in mouse, medaka, Xenopus, and zebrafish. To uncover novel Rx targets, we analyzed the expression of multiple eye development genes in chk. We find that expression of mab21l2, mab21l1 and rx2 are specifically absent in the eye field of chk embryos. Knockdown of Mab21l2 by antisense morpholino microinjections partially phenocopies the rx3 mutation, leading to microphthalmia, incomplete eye maturation, and dramatic increases in apoptotic eye progenitors. We propose that mab21l2 is an early downstream effector of rx3 and is critical for survival of eye progenitors.


Subject(s)
Eye/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Phenotype , Zebrafish/embryology , Animals , Chromosome Mapping , Crosses, Genetic , DNA, Complementary/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Microinjections , Morphogenesis , Mutation/genetics , Oligonucleotides , Polymorphism, Single-Stranded Conformational , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
16.
Proc Natl Acad Sci U S A ; 99(25): 16111-6, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12454286

ABSTRACT

Knockout of the murine retinoic acid (RA)-synthesizing enzyme retinaldehyde dehydrogenase 2 (RALDH2) gene leads to early morphogenetic defects and embryonic lethality. Using a RA-responsive reporter transgene, we have looked for RA-generating activities in Raldh2-null mouse embryos and investigated whether these activities could be ascribed to the other known RALDH enzymes (RALDH1 and RALDH3). To this end, the early defects of Raldh2(-/-) embryos were rescued through maternal dietary RA supplementation under conditions that do not interfere with the activity of the reporter transgene in WT embryos. We show that RALDH2 is responsible for most of the patterns of reporter transgene activity in the spinal cord and trunk mesodermal derivatives. However, reporter transgene activity was selectively detected in Raldh2(-/-) embryos within the mesonephric area that expresses RALDH3 and in medial-ventral cells of the spinal cord and posterior hindbrain, up to the level of the fifth rhombomere. The craniofacial patterns of RA-reporter activity were unaltered in Raldh2(-/-) mutants. Although these patterns correlated with the presence of Raldh1 andor Raldh3 transcripts in eye, nasal, and inner ear epithelia, no such correlation was found within forebrain neuroepithelium. These data suggest the existence of additional RA-generating activities in the differentiating forebrain, hindbrain, and spinal cord, which, along with RALDH1 and RALDH3, may account for the development of Raldh2(-/-) mutants once these have been rescued for early lethality.


Subject(s)
Aldehyde Oxidoreductases/physiology , Tretinoin/metabolism , Administration, Oral , Aldehyde Oxidoreductases/deficiency , Aldehyde Oxidoreductases/genetics , Animals , Ear, Inner/embryology , Ear, Inner/metabolism , Epithelial Cells/metabolism , Eye/embryology , Eye/metabolism , Female , Fetal Diseases/drug therapy , Gene Expression Regulation, Developmental , Genes, Lethal , Genes, Reporter , Gestational Age , Lac Operon , Mesonephros/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Nasal Mucosa/embryology , Nasal Mucosa/metabolism , Organ Specificity , Pregnancy , Prosencephalon/embryology , Prosencephalon/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Tretinoin/therapeutic use
17.
Hum Cell ; 15(3): 178-82, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12703548

ABSTRACT

Embryonic stem (ES) cells are pluripotent cells isolated from the inner cell mass of blastocysts. ES cells are able to differentiate into the three primitive layers (endoderm, mesoderm, and ectoderm) of the organism, including the germline. In recent reports mouse ES cells have been successfully applied in the treatment of spinal cord injury, hereditary myelin disorder of the central nervous system, and diabetes mellitus. In this study, we investigated the induction of mouse ES cell differentiation, using culture of embryoid bodies (EBs) into the diverse tissues. EBs were formed by culturing ES cells (129/SV strain) in DMEM supplemented with 10% FBS, in the absence of feeder cells and leukemia inhibitory factor (LF). EBs were induced to differentiate by treatment with retinoic acid (RA). In control medium (non-RA medium) beating muscles, blood vessels, hemocytes, and cartilages were frequently observed in EBs. Moreover, when EBs were cultured in medium including RA (5 x 10(-8) M, and 5 x 10(-9) M), differentiation of the optic vesicle, lens, retina, and neural groove was observed. In this study we demonstrated that an efficient system for inducing the differentiation of ES cells using EBs.


Subject(s)
Cell Differentiation/drug effects , Embryo, Mammalian/cytology , Mice/embryology , Stem Cells/cytology , Tretinoin/pharmacology , Animals , Cell Culture Techniques/methods , Culture Media , Eye/embryology , Morphogenesis/drug effects , Stimulation, Chemical
18.
Cell ; 107(4): 513-23, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11719191

ABSTRACT

In humans, low peak bone mass is a significant risk factor for osteoporosis. We report that LRP5, encoding the low-density lipoprotein receptor-related protein 5, affects bone mass accrual during growth. Mutations in LRP5 cause the autosomal recessive disorder osteoporosis-pseudoglioma syndrome (OPPG). We find that OPPG carriers have reduced bone mass when compared to age- and gender-matched controls. We demonstrate LRP5 expression by osteoblasts in situ and show that LRP5 can transduce Wnt signaling in vitro via the canonical pathway. We further show that a mutant-secreted form of LRP5 can reduce bone thickness in mouse calvarial explant cultures. These data indicate that Wnt-mediated signaling via LRP5 affects bone accrual during growth and is important for the establishment of peak bone mass.


Subject(s)
Bone Density/genetics , Eye Abnormalities/genetics , Eye/embryology , Osteoblasts/metabolism , Osteoporosis/genetics , Receptors, LDL/physiology , Transforming Growth Factor beta , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Adult , Animals , Animals, Outbred Strains , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Chromosomes, Human, Pair 11/genetics , Culture Media, Conditioned/pharmacology , DNA, Complementary/genetics , Dishevelled Proteins , Female , Genes, Recessive , Heterozygote , Humans , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Phosphoproteins/genetics , Phosphoproteins/physiology , Proteins/genetics , Proteins/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Fusion Proteins/physiology , Recombinant Proteins , Signal Transduction , Skull/cytology , Species Specificity , Stromal Cells/cytology , Stromal Cells/drug effects , Syndrome , Transfection , Wnt Proteins , Wnt-5a Protein , Wnt2 Protein , Wnt3 Protein , Wnt4 Protein
19.
Rev Med Chil ; 129(10): 1203-11, 2001 Oct.
Article in Spanish | MEDLINE | ID: mdl-11775350

ABSTRACT

Docosahexanoic acid (C22:6, DHA) is a highly unsaturated omega-3 fatty acid that forms part of the central nervous and visual system structures. DHA is synthesized from its precursor, alfa-linolenic acid, that is also a omega-3 fatty acid and can be obtained from vegetable oils. Marine organisms, specially fish, are good nutritional sources of DHA and eicosapentanoic acid (EPA), another omega-3 fatty acid that has a role in vascular homeostasis. DHA increases membrane fluidity, improving neurogenesis, synaptogenesis and the activity of retinal photoreceptors. The fetus, specially during the last trimester of pregnancy, has high DHA requirements. It is provided by the mother, since fetal DHA synthesis is negligible in this stage of development. Breast feeding provides DHA to the child, but most replacement artificial formulas do not provide this fatty acid. At the present moment, many products for infant nutrition contain DHA.


Subject(s)
Embryonic and Fetal Development/physiology , Fatty Acids, Omega-3/metabolism , Infant Nutritional Physiological Phenomena/physiology , Brain/embryology , Brain/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Embryonic and Fetal Development/drug effects , Endoplasmic Reticulum/metabolism , Eye/embryology , Eye/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6 , Fatty Acids, Unsaturated/metabolism , Female , Food, Fortified , Humans , Infant, Newborn , Pregnancy
20.
Mech Dev ; 99(1-2): 123-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11091080

ABSTRACT

Three homeobox genes, one from Drosophila melanogaster (Drosophila Hmx gene) and two from mouse (murine Hmx2 and Hmx3) were isolated and the full-length cDNAs and corresponding genomic structures were characterized. The striking homeodomain similarity encoded by these three genes to previously identified genes in sea urchin, chick and human, as well as the recently cloned murine Hmx1 gene, and the low homology to other homeobox genes indicate that the Hmx genes comprise a novel gene family. The widespread existence of Hmx genes in the animal kingdom suggests that this gene family is of ancient origin. Drosophila Hmx was mapped to the 90B5 region of Chromosome 3 and at early embryonic stages is primarily expressed in distinct areas of the neuroectoderm and subsets of neuroblasts in the developing fly brain. Later its expression continues in rostral areas of the brain in a segmented pattern, suggesting a putative role in the development of the Drosophila central nervous system. During evolution, mouse Hmx2 and Hmx3 may have retained a primary function in central nervous system development as suggested by their expression in the postmitotic cells of the neural tube, as well as in the hypothalamus, the mesencephalon, metencephalon and discrete regions in the myelencephalon during embryogenesis. Hmx1 has diverged from other Hmx members by its expression in the dorsal root, sympathetic and vagal nerve (X) ganglia. Aside from their expression in the developing nervous system, all three Hmx genes display expression in sensory organ development, and in the adult uterus. Hmx2 and Hmx3 show identical expression in the otic vesicle, whereas Hmx1 is strongly expressed in the developing eye. Transgenic mouse lines were generated to examine the DNA regulatory elements controlling Hmx2 and Hmx3. Transgenic constructs spanning more than 31 kb of genomic DNA gave reproducible expression patterns in the developing central and peripheral nervous systems, eye, ear and other tissues, yet failed to fully recapitulate the endogenous expression pattern of either Hmx2 or Hmx3, suggesting both the presence and absence of certain critical enhancers in the transgenes, or the requirement of proximal enhancers to work synergistically.


Subject(s)
Brain/embryology , Drosophila Proteins , Drosophila/embryology , Embryo, Mammalian/metabolism , Evolution, Molecular , Genes, Homeobox/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Transcription Factors , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary/metabolism , Drosophila/genetics , Ectoderm/metabolism , Embryo, Nonmammalian/metabolism , Eye/embryology , Ganglia/embryology , Gene Library , Humans , Hypothalamus/embryology , In Situ Hybridization , Mesencephalon/embryology , Metencephalon/embryology , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Nervous System/embryology , Neural Crest/embryology , RNA/metabolism , Sequence Homology, Amino Acid , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL