Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Eye Res ; 93(4): 460-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21722635

ABSTRACT

The zebrafish lens opaque (lop) mutant was previously isolated in a genetic screen and shown to lack rod and cone photoreceptors and exhibit lens opacity, or cataract, at 7 days post-fertilization (dpf). In this manuscript, we provide four different lines of evidence demonstrating that the lop phenotype results from a defect in the cdipt (phosphatidylinositol (PI) synthase; CDP-diacylglycerol-inositol 3-phosphatidyltransferase) gene. First, DNA sequence analysis revealed that the lop mutant contained a missense mutation in the lop open reading frame, which yields a nonconservative amino acid substitution (Ser-111-Cys) within the PI synthase catalytic domain. Second, morpholino-mediated knockdown of the cdipt-encoded PI synthase protein phenocopied the cdipt(lop/lop) mutant, with abnormal lens epithelial and secondary fiber cell morphologies and reduced numbers of photoreceptors. Third, microinjection of in vitro transcribed, wild-type cdipt mRNA into 1-4 cell stage cdipt(lop/lop) embryos significantly reduced the percentage of larvae displaying lens opacity at 7 dpf. Fourth, a cdipt retroviral-insertion allele, cdipt(hi559), exhibited similar lens and retinal abnormalities and failed to complement the cdipt(lop) mutant phenotype. To determine the initial cellular defects associated with the cdipt mutant, we examined homozygous cdipt(hi559/hi559) mutants prior to gross lens opacification at 6 dpf. The cdipt(hi559/hi559) mutants first exhibited photoreceptor layer disruption and photoreceptor cell death at 3 and 4 dpf, respectively, followed by lens dismorphogenesis by 5 dpf. RT-PCR revealed that the cdipt gene is maternally expressed and continues to be transcribed throughout development and into adulthood, in a wide variety of tissues. Using an anti-zebrafish PI synthase polyclonal antiserum, we localized the protein throughout the developing eye, including the photoreceptor layer and lens cortical secondary fiber cells. As expected, the polyclonal antiserum revealed that the PI synthase protein was reduced in amount in both the cdipt(lop/lop) and cdipt(hi559/hi559) mutants. Furthermore, we used a heterologous yeast phenotypic complementation assay to confirm that the wild-type zebrafish cdipt allele encodes functional PI synthase activity. Taken together, the cdipt-encoded PI synthase is required for survival of photoreceptor cells and lens epithelial and secondary cortical fiber cells. These zebrafish cdipt alleles represent excellent in vivo genetic tools to study the role of phosphatidylinositol and its phosphorylated derivatives in lens and photoreceptor development and maintenance.


Subject(s)
CDP-Diacylglycerol-Inositol 3-Phosphatidyltransferase/physiology , Lens, Crystalline/cytology , Membrane Proteins/physiology , Photoreceptor Cells, Vertebrate/cytology , Zebrafish Proteins/physiology , Animals , Apoptosis , Cataract/genetics , Cell Survival/physiology , DNA Primers/chemistry , Epithelial Cells/cytology , Epithelial Cells/enzymology , Fluorescent Antibody Technique, Indirect , Genotype , Immunoblotting , In Situ Nick-End Labeling , Lens, Crystalline/enzymology , MicroRNAs/genetics , Mutation, Missense , Photoreceptor Cells, Vertebrate/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish
2.
PLoS Genet ; 7(2): e1001310, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21379331

ABSTRACT

The glaucomas comprise a genetically complex group of retinal neuropathies that typically occur late in life and are characterized by progressive pathology of the optic nerve head and degeneration of retinal ganglion cells. In addition to age and family history, other significant risk factors for glaucoma include elevated intraocular pressure (IOP) and myopia. The complexity of glaucoma has made it difficult to model in animals, but also challenging to identify responsible genes. We have used zebrafish to identify a genetically complex, recessive mutant that shows risk factors for glaucoma including adult onset severe myopia, elevated IOP, and progressive retinal ganglion cell pathology. Positional cloning and analysis of a non-complementing allele indicated that non-sense mutations in low density lipoprotein receptor-related protein 2 (lrp2) underlie the mutant phenotype. Lrp2, previously named Megalin, functions as an endocytic receptor for a wide-variety of bioactive molecules including Sonic hedgehog, bone morphogenic protein 4, retinol-binding protein, vitamin D-binding protein, and apolipoprotein E, among others. Detailed phenotype analyses indicated that as lrp2 mutant fish age, many individuals--but not all--develop high IOP and severe myopia with obviously enlarged eye globes. This results in retinal stretch and prolonged stress to retinal ganglion cells, which ultimately show signs of pathogenesis. Our studies implicate altered Lrp2-mediated homeostasis as important for myopia and other risk factors for glaucoma in humans and establish a new genetic model for further study of phenotypes associated with this disease.


Subject(s)
Eye/pathology , Glaucoma/complications , Glaucoma/genetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mutation/genetics , Myopia/complications , Myopia/genetics , Zebrafish Proteins/genetics , Aging/pathology , Amino Acid Sequence , Animals , Apoptosis , Axons/pathology , Base Sequence , Cell Count , Cell Proliferation , Disease Models, Animal , Glaucoma/physiopathology , Hydrophthalmos/complications , Intraocular Pressure , Low Density Lipoprotein Receptor-Related Protein-2/chemistry , Molecular Sequence Data , Myopia/physiopathology , Optic Disk/pathology , Optic Disk/ultrastructure , Organ Size , Phenotype , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Risk Factors , Stress, Physiological/genetics , Up-Regulation , Zebrafish/genetics , Zebrafish Proteins/chemistry
3.
Invest Ophthalmol Vis Sci ; 52(6): 3119-28, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21330665

ABSTRACT

PURPOSE: To establish the zebrafish platinum mutant as a model for studying vision defects caused by syndromic albinism diseases such as Chediak-Higashi syndrome, Griscelli syndrome, and Hermansky-Pudlak syndrome (HPS). METHODS: Bulked segregant analysis and candidate gene sequencing revealed that the zebrafish platinum mutation is a single-nucleotide insertion in the vps11 (vacuolar protein sorting 11) gene. Expression of vps11 was determined by RT-PCR and in situ hybridization. Mutants were analyzed for pigmentation defects and retinal disease by histology, immunohistochemistry, and transmission electron microscopy. RESULTS: Phenocopy and rescue experiments determined that a loss of Vps11 results in the platinum phenotype. Expression of vps11 appeared ubiquitous during zebrafish development, with stronger expression in the developing retina and retinal pigmented epithelium (RPE). Zebrafish platinum mutants exhibited reduced pigmentation in the body and RPE; however, melanophore development, migration, and dispersion occurred normally. RPE, photoreceptors, and inner retinal neurons formed normally in zebrafish platinum mutants. However, a gradual loss of RPE, an absence of mature melanosomes, and the subsequent degradation of RPE/photoreceptor interdigitation was observed. CONCLUSIONS: These data show that Vps11 is not necessary for normal retinal development or initiation of melanin biosynthesis, but is essential for melanosome maturation and healthy maintenance of the RPE and photoreceptors.


Subject(s)
Albinism, Oculocutaneous/genetics , Disease Models, Animal , Polymorphism, Single Nucleotide , Retinal Diseases/genetics , Vesicular Transport Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/genetics , Albinism, Oculocutaneous/metabolism , Albinism, Oculocutaneous/pathology , Animals , Chediak-Higashi Syndrome/genetics , Fluorescent Antibody Technique, Indirect , Hearing Loss, Sensorineural/genetics , Hepatomegaly/genetics , Hermanski-Pudlak Syndrome/genetics , In Situ Hybridization , Melanins/biosynthesis , Melanophores/metabolism , Melanosomes/genetics , Melanosomes/metabolism , Microscopy, Electron, Transmission , Models, Genetic , Mutation , Pericardial Effusion/genetics , Piebaldism/genetics , Pigmentation Disorders/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Reverse Transcriptase Polymerase Chain Reaction , Syndrome , Zebrafish/embryology
4.
PLoS One ; 5(10): e13743, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-21060795

ABSTRACT

Congenital disorder of glycosylation type IIc (CDG IIc) is characterized by mental retardation, slowed growth and severe immunodeficiency, attributed to the lack of fucosylated glycoproteins. While impaired Notch signaling has been implicated in some aspects of CDG IIc pathogenesis, the molecular and cellular mechanisms remain poorly understood. We have identified a zebrafish mutant slytherin (srn), which harbors a missense point mutation in GDP-mannose 4,6 dehydratase (GMDS), the rate-limiting enzyme in protein fucosylation, including that of Notch. Here we report that some of the mechanisms underlying the neural phenotypes in srn and in CGD IIc are Notch-dependent, while others are Notch-independent. We show, for the first time in a vertebrate in vivo, that defects in protein fucosylation leads to defects in neuronal differentiation, maintenance, axon branching, and synapse formation. Srn is thus a useful and important vertebrate model for human CDG IIc that has provided new insights into the neural phenotypes that are hallmarks of the human disorder and has also highlighted the role of protein fucosylation in neural development.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Disease Models, Animal , Hydro-Lyases/genetics , Nervous System/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Glycosylation , Humans , In Situ Hybridization , Molecular Sequence Data , Mutation, Missense , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Zebrafish
5.
PLoS One ; 4(12): e8329, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-20020044

ABSTRACT

In humans, mutations in electron transfer flavoprotein (ETF) or electron transfer flavoprotein dehydrogenase (ETFDH) lead to MADD/glutaric aciduria type II, an autosomal recessively inherited disorder characterized by a broad spectrum of devastating neurological, systemic and metabolic symptoms. We show that a zebrafish mutant in ETFDH, xavier, and fibroblast cells from MADD patients demonstrate similar mitochondrial and metabolic abnormalities, including reduced oxidative phosphorylation, increased aerobic glycolysis, and upregulation of the PPARG-ERK pathway. This metabolic dysfunction is associated with aberrant neural proliferation in xav, in addition to other neural phenotypes and paralysis. Strikingly, a PPARG antagonist attenuates aberrant neural proliferation and alleviates paralysis in xav, while PPARG agonists increase neural proliferation in wild type embryos. These results show that mitochondrial dysfunction, leading to an increase in aerobic glycolysis, affects neurogenesis through the PPARG-ERK pathway, a potential target for therapeutic intervention.


Subject(s)
Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Nervous System/metabolism , Nervous System/pathology , Zebrafish/metabolism , Animals , Carboxylic Acids/metabolism , Carnitine/analogs & derivatives , Carnitine/blood , Cell Proliferation/drug effects , Cloning, Molecular , Electron-Transferring Flavoproteins/genetics , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Glycolysis/drug effects , Humans , Infant , Infant, Newborn , Iron-Sulfur Proteins/genetics , Mitochondria/drug effects , Mitochondria/pathology , Mutation/genetics , Nervous System/drug effects , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Oligonucleotides, Antisense/pharmacology , Oxidoreductases Acting on CH-NH Group Donors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Phenotype
6.
J Neurosci ; 29(42): 13190-201, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19846707

ABSTRACT

Multiple molecular cues guide neuronal axons to their targets during development. Previous studies in vitro have shown that mechanical stimulation also can affect axon growth; however, whether mechanical force contributes to axon guidance in vivo is unknown. We investigated the role of muscle contractions in the guidance of zebrafish peripheral Rohon-Beard (RB) sensory axons in vivo. We analyzed several mutants that affect muscle contraction through different molecular pathways, including a new mutant allele of the titin a (pik) gene, mutants that affect the hedgehog signaling pathway, and a nicotinic acetylcholine receptor mutant. We found RB axon defects in these mutants, the severity of which appeared to correlate with the extent of muscle contraction loss. These axons extend between the muscle and skin and normally have ventral trajectories and repel each other on contact. RB peripheral axons in muscle mutants extend longitudinally instead of ventrally, and the axons fail to repel one another on contact. In addition, we showed that limiting muscle movements by embedding embryos in agarose caused similar defects in peripheral RB axon guidance. This work suggests that the mechanical forces generated by muscle contractions are necessary for proper sensory axon pathfinding in vivo.


Subject(s)
Axons/physiology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Peripheral Nerves/cytology , Sensory Receptor Cells/cytology , Animals , Animals, Genetically Modified , Axons/drug effects , Body Patterning/drug effects , Body Patterning/genetics , Chondroitin ABC Lyase/pharmacology , Connectin , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Hedgehog Proteins/metabolism , Muscle Contraction/drug effects , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/embryology , Mutation/genetics , Neuromuscular Nondepolarizing Agents/pharmacology , Protein Kinases/genetics , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors , Tubocurarine/pharmacology , Zebrafish , Zebrafish Proteins/genetics
7.
Dev Biol ; 319(1): 10-22, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18479681

ABSTRACT

In this study, we have characterized the ocular defects in the recessive zebrafish mutant blowout that presents with a variably penetrant coloboma phenotype. blowout mutants develop unilateral or bilateral colobomas and as a result, the retina and retinal pigmented epithelium are not contained within the optic cup. Colobomas result from defects in optic stalk morphogenesis whereby the optic stalk extends into the retina and impedes the lateral edges of the choroid fissure from meeting and fusing. The expression domain of the proximal optic vesicle marker pax2a is expanded in blowout at the expense of the distal optic vesicle marker pax6, suggesting that the initial patterning of the optic vesicle into proximal and distal territories is disrupted in blowout. Later aspects of distal optic cup formation (i.e. retina development) are normal in blowout mutants, however. Positional cloning of blowout identified a nonsense mutation in patched1, a negative regulator of the Hedgehog pathway, as the underlying cause of the blowout phenotype. Expanded domains of expression of the Hedgehog target genes patched1 and patched2 were observed in blowout, consistent with a loss of Patched1 function and upregulation of Hedgehog pathway activity. Moreover, colobomas in blowout could be suppressed by pharmacologically inhibiting the Hedgehog pathway with cyclopamine, and maximal rescue occurred when embryos were exposed to cyclopamine between 5.5 and 13 hours post-fertilization. These observations highlight the critical role that Hedgehog pathway activity plays in mediating patterning of the proximal/distal axis of the optic vesicle during the early phases of eye development and they provide genetic confirmation for the integral role that patched1-mediated negative regulation of Hedgehog signaling plays during vertebrate eye development.


Subject(s)
Coloboma/embryology , Eye/embryology , Hedgehog Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Body Patterning , Choroid/embryology , Coloboma/metabolism , Embryo, Nonmammalian/metabolism , Eye/metabolism , Membrane Proteins/metabolism , Patched Receptors , Patched-1 Receptor , Signal Transduction , Zebrafish , Zebrafish Proteins/metabolism
8.
Dev Biol ; 299(1): 63-77, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16973147

ABSTRACT

We report phenotypic and genetic analyses of a recessive, larval lethal zebrafish mutant, bal(a69), characterized by severe eye defects and shortened body axis. The bal(a69) mutation was mapped to chromosome 24 near the laminin alpha 1 (lama1) gene. We analyzed the lama1 gene sequence within bal(a69) embryos and two allelic mutants, bal(arl) and bal(uw1). Missense (bal(a69)), nonsense (bal(arl)), and frameshift (bal(uw1)) alterations in lama1 were found to underlie the phenotypes. Extended analysis of bal(a69) ocular features revealed disrupted lens development with subsequent lens degeneration, focal cornea dysplasia, and hyaloid vasculature defects. Within the neural retina, the ganglion cells showed axonal projection defects and ectopic photoreceptor cells were noted at inner retinal locations. To address whether ocular anomalies were secondary to defects in lens differentiation, bal(a69) mutants were compared to embryos in which the lens vesicle was surgically removed. Our analysis suggests that many of the anterior and posterior ocular defects in bal(a69) are independent of the lens degeneration. Analysis of components of focal adhesion signaling complexes suggests that reduced focal adhesion kinase activation underlies the anterior segment dysgenesis in lama1 mutants. To assess adult ocular phenotypes associated with lama1 mutations, genetic mosaics were generated by transplanting labeled bal cells into ocular-fated regions of wild-type blastulas. Adult chimeric eyes displayed a range of defects including anterior segment dysgenesis and cataracts. Our analysis provides mechanistic insights into the developmental defects and ocular pathogenesis caused by mutations in laminin subunits.


Subject(s)
Laminin/deficiency , Laminin/genetics , Lens, Crystalline/embryology , Mutation/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Anterior Eye Segment/abnormalities , Anterior Eye Segment/cytology , Anterior Eye Segment/embryology , Base Sequence , Cataract/pathology , Chromosomes/genetics , Cloning, Molecular , DNA Mutational Analysis , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Focal Adhesions , Gene Expression Regulation, Developmental , Laminin/chemistry , Lens, Crystalline/abnormalities , Lens, Crystalline/cytology , Lens, Crystalline/surgery , Molecular Sequence Data , Mosaicism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Ganglion Cells/cytology , Retinal Vessels/abnormalities , Retinal Vessels/embryology , Zebrafish/abnormalities
9.
Proc Natl Acad Sci U S A ; 100(11): 6535-40, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12748389

ABSTRACT

Zebrafish with the young (yng) mutation show a defect in retinal cell differentiation. Here we demonstrate that a mutation in a brahma-related gene (brg1) is responsible for the yng phenotype. Brahma homologues function as essential subunits for SWI/SNF-type chromatin remodeling complexes. Our analysis indicates that brg1 is required for the wave of mitogen-activated protein kinase activity that precedes retinal cell differentiation. Using specific inhibitors of the mitogen-activated protein kinase pathway we show this signal has a direct role in retinal cell differentiation. Lastly, through investigations of mutants in other chromatin remodeling subunits, we provide genetic evidence for gene and tissue specificity of the Brahma chromatin remodeling complex.


Subject(s)
Cell Cycle Proteins/genetics , Chromatin/metabolism , Mutation , Trans-Activators/genetics , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Drosophila Proteins , Molecular Sequence Data , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...