Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Publication year range
1.
Hum Mutat ; 41(3): 678-695, 2020 03.
Article in English | MEDLINE | ID: mdl-31816153

ABSTRACT

Uveal coloboma is a potentially blinding congenital ocular malformation caused by the failure of optic fissure closure during the fifth week of human gestation. We performed custom capture high-throughput screening of 38 known coloboma-associated genes in 66 families. Suspected causative novel variants were identified in TFAP2A and CHD7, as well as two previously reported variants of uncertain significance in RARB and BMP7. The variant in RARB, unlike previously reported disease mutations in the ligand-binding domain, was a missense change in the highly conserved DNA-binding domain predicted to affect the protein's DNA-binding ability. In vitro studies revealed lower steady-state protein levels, reduced transcriptional activity, and incomplete nuclear localization of the mutant RARB protein compared with wild-type. Zebrafish studies showed that human RARB messenger RNA partially reduced the ocular phenotype caused by morpholino knockdown of rarga gene, a zebrafish homolog of human RARB. Our study indicates that sequence alterations in known coloboma genes account for a small percentage of coloboma cases and that mutations in the RARB DNA-binding domain could result in human disease.


Subject(s)
Coloboma/diagnosis , Coloboma/genetics , DNA-Binding Proteins/metabolism , High-Throughput Nucleotide Sequencing , Mutation , Protein Interaction Domains and Motifs , Receptors, Retinoic Acid/metabolism , Adult , Animals , Child , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , Female , Gene Expression , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Humans , Infant , Male , Models, Molecular , Pedigree , Phenotype , Receptors, Retinoic Acid/chemistry , Structure-Activity Relationship , Zebrafish
2.
Exp Eye Res ; 193: 107940, 2020 04.
Article in English | MEDLINE | ID: mdl-32032630

ABSTRACT

Optic fissure closure defects result in uveal coloboma, a potentially blinding condition affecting between 0.5 and 2.6 per 10,000 births that may cause up to 10% of childhood blindness. Uveal coloboma is on a phenotypic continuum with microphthalmia (small eye) and anophthalmia (primordial/no ocular tissue), the so-called MAC spectrum. This review gives a brief overview of the developmental biology behind coloboma and its clinical presentation/spectrum. Special attention will be given to two prominent, syndromic forms of coloboma, namely, CHARGE (Coloboma, Heart defect, Atresia choanae, Retarded growth and development, Genital hypoplasia, and Ear anomalies/deafness) and COACH (Cerebellar vermis hypoplasia, Oligophrenia, Ataxia, Coloboma, and Hepatic fibrosis) syndromes. Approaches employed to identify genes involved in optic fissure closure in animal models and recent advances in live imaging of zebrafish eye development are also discussed.


Subject(s)
Abnormalities, Multiple/genetics , Ataxia/genetics , Brain/abnormalities , Cholestasis/genetics , Coloboma/genetics , Genetic Predisposition to Disease , Liver Diseases/genetics , Uvea/abnormalities , Animals , Humans
3.
Stem Cells ; 31(6): 1149-59, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23495178

ABSTRACT

Replacement of dysfunctional or dying photoreceptors offers a promising approach for retinal neurodegenerative diseases, including age-related macular degeneration and retinitis pigmentosa. Several studies have demonstrated the integration and differentiation of developing rod photoreceptors when transplanted in wild-type or degenerating retina; however, the physiology and function of the donor cells are not adequately defined. Here, we describe the physiological properties of developing rod photoreceptors that are tagged with green fluorescent protein (GFP) driven by the promoter of rod differentiation factor, Nrl. GFP-tagged developing rods show Ca(2 +) responses and rectifier outward currents that are smaller than those observed in fully developed photoreceptors, suggesting their immature developmental state. These immature rods also exhibit hyperpolarization-activated current (Ih ) induced by the activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. When transplanted into the subretinal space of wild-type or retinal degeneration mice, GFP-tagged developing rods can integrate into the photoreceptor outer nuclear layer in wild-type mouse retina and exhibit Ca(2 +) responses and membrane current comparable to native rod photoreceptors. A proportion of grafted rods develop rhodopsin-positive outer segment-like structures within 2 weeks after transplantation into the retina of Crx-knockout mice and produce rectifier outward current and Ih upon membrane depolarization and hyperpolarization. GFP-positive rods derived from induced pluripotent stem (iPS) cells also display similar membrane current Ih as native developing rod photoreceptors, express rod-specific phototransduction genes, and HCN-1 channels. We conclude that Nrl-promoter-driven GFP-tagged donor photoreceptors exhibit physiological characteristics of rods and that iPS cell-derived rods in vitro may provide a renewable source for cell-replacement therapy.


Subject(s)
Homeodomain Proteins/genetics , Photoreceptor Cells, Vertebrate/physiology , Retina/physiology , Retinal Degeneration/therapy , Retinal Rod Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/transplantation , Trans-Activators/genetics , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Cell Differentiation/genetics , Cells, Cultured , Eye Proteins/genetics , Eye Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/metabolism , Promoter Regions, Genetic , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/metabolism , Stem Cell Transplantation , Trans-Activators/metabolism
4.
Hum Genomics ; 7: 13, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23676014

ABSTRACT

The Kelch-like (KLHL) gene family encodes a group of proteins that generally possess a BTB/POZ domain, a BACK domain, and five to six Kelch motifs. BTB domains facilitate protein binding and dimerization. The BACK domain has no known function yet is of functional importance since mutations in this domain are associated with disease. Kelch domains form a tertiary structure of ß-propellers that have a role in extracellular functions, morphology, and binding to other proteins. Presently, 42 KLHL genes have been classified by the HUGO Gene Nomenclature Committee (HGNC), and they are found across multiple human chromosomes. The KLHL family is conserved throughout evolution. Phylogenetic analysis of KLHL family members suggests that it can be subdivided into three subgroups with KLHL11 as the oldest member and KLHL9 as the youngest. Several KLHL proteins bind to the E3 ligase cullin 3 and are known to be involved in ubiquitination. KLHL genes are responsible for several Mendelian diseases and have been associated with cancer. Further investigation of this family of proteins will likely provide valuable insights into basic biology and human disease.


Subject(s)
Carrier Proteins/genetics , Genetic Diseases, Inborn/genetics , Multigene Family/genetics , Neoplasms/genetics , Repetitive Sequences, Amino Acid/genetics , Carrier Proteins/classification , Carrier Proteins/metabolism , Humans , Neoplasms/etiology , Phylogeny , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Ubiquitination/genetics
5.
Ophthalmic Genet ; 44(2): 182-185, 2023 04.
Article in English | MEDLINE | ID: mdl-36951427

ABSTRACT

BACKGROUND: Variations in the protocadherin gene FAT1 have recently been associated with a syndrome that includes coloboma, facial dysmorphism, renal failure, syndactyly, and other developmental defects. MATERIALS AND METHODS: Detailed medical and family history, physical examination, and molecular analysis. RESULTS: This non-dysmorphic, intellectually normal 51-year-old woman presented with bilateral colobomata and renal failure of unclear etiology, and asymmetric sensorineural hearing loss. Family history was notable for multiple family members with various forms of cancer. Whole exome sequencing revealed a homozygous frame shift variant in FAT1, predicted to truncate the FAT1 protein at the furthest position in the protein structure published to date in a patient with coloboma. CONCLUSIONS: This case provides further evidence of the pleiotropic effects of FAT1 in optic fissure closure and kidney function. Also, because this variant is in the last exon, it would be anticipated to escape nonsense-mediated decay, opening the possibility that the protein is made and expressed, but not completely functional, as its intracellular domain is truncated.


Subject(s)
Coloboma , Renal Insufficiency , Female , Humans , Middle Aged , Coloboma/diagnosis , Coloboma/genetics , Protocadherins , Cadherins/genetics
6.
Pharmacol Ther ; 249: 108482, 2023 09.
Article in English | MEDLINE | ID: mdl-37385300

ABSTRACT

Stargardt maculopathy, caused predominantly by mutations in the ABCA4 gene, is characterized by an accumulation of non-degradable visual pigment derivative, lipofuscin, in the retinal pigment epithelium (RPE) - resulting in RPE atrophy. RPE is a monolayer tissue located adjacent to retinal photoreceptors and regulates their health and functioning; RPE atrophy triggers photoreceptor cell death and vision loss in Stargardt patients. Previously, ABCA4 mutations in photoreceptors were thought to be the major contributor to lipid homeostasis defects in the eye. Recently, we demonstrated that ABCA4 loss of function in the RPE leads to cell-autonomous lipid homeostasis defects. Our work underscores that an incomplete understanding of lipid metabolism and lipid-mediated signaling in the retina and RPE are potential causes for lacking treatments for this disease. Here we report altered lipidomic in mouse and human Stargardt models. This work provides the basis for therapeutics that aim to restore lipid homeostasis in the retina and the RPE.


Subject(s)
Macular Degeneration , Retinal Degeneration , Humans , Mice , Animals , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retina/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Lipofuscin/genetics , Lipofuscin/metabolism , Atrophy/metabolism , Atrophy/pathology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism
7.
Stem Cell Reports ; 17(1): 173-186, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021041

ABSTRACT

Oculocutaneous albinism (OCA) encompasses a set of autosomal recessive genetic conditions that affect pigmentation in the eye, skin, and hair. OCA patients display reduced best-corrected visual acuity, reduced to absent ocular pigmentation, abnormalities in fovea development, and/or abnormal decussation of optic nerve fibers. It has been hypothesized that improving eye pigmentation could prevent or rescue some of the vision defects. The goal of the present study was to develop an in vitro model for studying pigmentation defects in human retinal pigment epithelium (RPE). We developed a "disease in a dish" model for OCA1A and OCA2 types using induced pluripotent stem cells to generate RPE. The RPE is a monolayer of cells that are pigmented, polarized, and polygonal in shape, located between the neural retina and choroid, with an important role in vision. Here we show that RPE tissue derived in vitro from OCA patients recapitulates the pigmentation defects seen in albinism, while retaining the apical-basal polarity and normal polygonal morphology of the constituent RPE cells.


Subject(s)
Albinism, Oculocutaneous/etiology , Albinism, Oculocutaneous/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium/metabolism , Albinism, Oculocutaneous/pathology , Animals , Biomarkers , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Humans , Melanocytes/metabolism , Melanocytes/ultrastructure , Phenotype , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/ultrastructure
8.
Invest Ophthalmol Vis Sci ; 63(12): 5, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36326727

ABSTRACT

Purpose: Uveal coloboma is a congenital eye malformation caused by failure of the optic fissure to close in early human development. Despite significant progress in identifying genes whose regulation is important for executing this closure, mutations are detected in a minority of cases using known gene panels, implying additional genetic complexity. We have previously shown knockdown of znf503 (the ortholog of mouse Zfp503) in zebrafish causes coloboma. Here we characterize Zfp503 knockout (KO) mice and evaluate transcriptomic profiling of mutant versus wild-type (WT) retinal pigment epithelium (RPE)/choroid. Methods: Zfp503 KO mice were generated by gene targeting using homologous recombination. Embryos were characterized grossly and histologically. Patterns and level of developmentally relevant proteins/genes were examined with immunostaining/in situ hybridization. The transcriptomic profile of E11.5 KO RPE/choroid was compared to that of WT. Results: Zfp503 is dynamically expressed in developing mouse eyes, and loss of its expression results in uveal coloboma. KO embryos exhibit altered mRNA levels and expression patterns of several key transcription factors involved in eye development, including Otx2, Mitf, Pax6, Pax2, Vax1, and Vax2, resulting in a failure to maintain the presumptive RPE, as evidenced by reduced melanin pigmentation and its differentiation into a neural retina-like lineage. Comparison of RNA sequencing data from WT and KO E11.5 embryos demonstrated reduced expression of melanin-related genes and significant overlap with genes known to be dynamically regulated at the optic fissure. Conclusions: These results demonstrate a critical role of Zfp503 in maintaining RPE fate and optic fissure closure.


Subject(s)
Coloboma , Neuropeptides , Animals , Humans , Mice , Coloboma/genetics , Coloboma/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Zebrafish/genetics
9.
Mol Vis ; 17: 2580-95, 2011.
Article in English | MEDLINE | ID: mdl-22025893

ABSTRACT

PURPOSE: Cell replacement has the potential to be applied as a therapeutic strategy in retinal degenerative diseases such as retinitis pigmentosa and age-related macular degeneration (AMD) for which no adequate pharmacological and surgical treatments are currently available. Although controversial, the use of ciliary epithelium (CE)-derived cells is supported by evidence showing their differentiation into retinal phenotypes. This study examines the differentiation potential of porcine CE-derived cells in vitro and their survival, migration, morphological characteristics, and immunohistochemical phenotype in vivo, upon transplantation into the subretinal space of normal pigs. METHODS: Cells were isolated from the CE of postnatal pigs and were grown in a suspension sphere culture. Differentiation was assessed in vitro after exposure to laminin and the addition of serum. For transplantation, CE-derived spheres were dissociated, labeled with CM-DiI vital dye, and the cells were injected subretinally into one eye of eight week-old allorecipients. The eyes were examined at eight days and at two and four weeks after transplantation. RESULTS: Cells positive for neuronal and retinal pigment epithelium (RPE) markers were detected by immunohistochemistry in differentiation cultures. Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR) revealed upregulation of neuronal markers after in vitro differentiation. CM-DiI dye-labeled CE-derived cells dissociated from primary spheres survived for up to four weeks after transplantation in vivo. Some of the surviving cells migrated distantly from the injection site. Large clusters of transplanted cells integrated into the RPE layer and multilayered RPE-like structures positive for RPE65 were often observed. Grafted cells were also identified in the neuroretina where 5%-10% were positive for recoverin, protein kinase C alpha (PKCα), and calbindin. CONCLUSIONS: The efficient conversion to an RPE-like phenotype suggests that CE-derived cells could be a potential source of RPE for cell replacement. Our data also suggest that the ability of these cells to acquire neuronal phenotypes is influenced by the environment. Thus, pre-differentiated or (re)programmed CE-derived cells may be more amenable for retinal repair.


Subject(s)
Cell Transplantation/methods , Epithelial Cells/cytology , Injections, Intraocular/methods , Neurons/cytology , Retinal Pigment Epithelium/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Carbocyanines/analysis , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Eye Proteins/metabolism , Fluorescent Dyes/analysis , Immunohistochemistry , Laminin/pharmacology , Macular Degeneration/pathology , Macular Degeneration/therapy , Neurons/metabolism , Retinal Pigment Epithelium/growth & development , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/therapy , Reverse Transcriptase Polymerase Chain Reaction , Swine , Transplantation, Homologous , cis-trans-Isomerases
10.
BMC Dev Biol ; 10: 1, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053268

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small RNA molecules (~22 nucleotides) which have been shown to play an important role both in development and in maintenance of adult tissue. Conditional inactivation of miRNAs in the eye causes loss of visual function and progressive retinal degeneration. In addition to inhibiting translation, miRNAs can mediate degradation of targeted mRNAs. We have previously shown that candidate miRNAs affecting transcript levels in a tissue can be deduced from mRNA microarray expression profiles. The purpose of this study was to predict miRNAs which affect mRNA levels in developing and adult retinal tissue and to confirm their expression. RESULTS: Microarray expression data from ciliary epithelial retinal stem cells (CE-RSCs), developing and adult mouse retina were generated or downloaded from public repositories. Analysis of gene expression profiles detected the effects of multiple miRNAs in CE-RSCs and retina. The expression of 20 selected miRNAs was confirmed by RT-PCR and the cellular distribution of representative candidates analyzed by in situ hybridization. The expression levels of miRNAs correlated with the significance of their predicted effects upon mRNA expression. Highly expressed miRNAs included miR-124, miR-125a, miR-125b, miR-204 and miR-9. Over-expression of three miRNAs with significant predicted effects upon global mRNA levels resulted in a decrease in mRNA expression of five out of six individual predicted target genes assayed. CONCLUSIONS: This study has detected the effect of miRNAs upon mRNA expression in immature and adult retinal tissue and cells. The validity of these observations is supported by the experimental confirmation of candidate miRNA expression and the regulation of predicted target genes following miRNA over-expression. Identified miRNAs are likely to be important in retinal development and function. Misregulation of these miRNAs might contribute to retinal degeneration and disease. Conversely, manipulation of their expression could potentially be used as a therapeutic tool in the future.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Retina/growth & development , Animals , Gene Expression Profiling , Mice , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Retina/metabolism
11.
Invest Ophthalmol Vis Sci ; 61(12): 25, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33107904

ABSTRACT

Purpose: Disruption of proteostasis is a key event in many neurodegenerative diseases. Heat shock proteins (HSPs) participate in multiple functions associated with intracellular transport and proteostasis. We evaluated the effect of augmented HSP70 expression in mutant photoreceptors of mouse retinal degeneration models to test the hypothesis that failure to sustain HSP70 expression contributes to photoreceptor cell death. Methods: We examined HSP70 expression in retinas of wild-type and mutant mice by RNA and protein analysis. A transgenic mouse line, TgCrx-Hspa1a-Flag, was generated to express FLAG-tagged full-length HSP70 protein under control of a 2.3 kb mouse Crx promoter. This line was crossed to three distinct retinal degeneration mouse models. Retinal structure and function were evaluated by histology, immunohistochemistry, and electroretinography. Results: In seven different mouse models of retinal degeneration, we detected transient elevation of endogenous HSP70 expression at early stages, followed by a dramatic reduction as cell death ensues, suggesting an initial adaptive response to cellular stress. Augmented expression of HSP70 in RHOT17M mice, in which mutant rhodopsin is misfolded, marginally improved photoreceptor survival, whereas elevated HSP70 led to more severe retinal degeneration in rd10 mutants that produce a partially functional PDE6B. In Rpgrip1-/- mice that display a ciliary defect, higher HSP70 had no impact on photoreceptor survival or function. Conclusions: HSP70 overexpression has divergent effects in photoreceptors determined, at least in part, by the nature of the mutant protein each model carries. Additional investigations on HSP pathways and associated chaperone networks in photoreceptors are needed before designing therapeutic strategies targeting proteostasis.


Subject(s)
Gene Expression Regulation/physiology , HSP70 Heat-Shock Proteins/genetics , Retinal Degeneration/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Cell Survival , Disease Models, Animal , Electroretinography , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Retina/physiopathology , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/physiology
12.
Cell Rep ; 31(3): 107525, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320661

ABSTRACT

Aging-associated functional decline is accompanied by alterations in the epigenome. To explore DNA modifications that could influence visual function with age, we perform whole-genome bisulfite sequencing of purified mouse rod photoreceptors at four ages and identify 2,054 differentially methylated regions (DMRs). We detect many DMRs during early stages of aging and in rod regulatory regions, and some of these cluster at chromosomal hotspots, especially on chromosome 10, which includes a longevity interactome. Integration of methylome to age-related transcriptome changes, chromatin signatures, and first-order protein-protein interactions uncover an enrichment of DMRs in altered pathways that are associated with rod function, aging, and energy metabolism. In concordance, we detect reduced basal mitochondrial respiration and increased fatty acid dependency with retinal age in ex vivo assays. Our study reveals age-dependent genomic and chromatin features susceptible to DNA methylation changes in rod photoreceptors and identifies a link between DNA methylation and energy metabolism in aging.


Subject(s)
Aging/genetics , DNA Methylation/genetics , Energy Metabolism/genetics , Retinal Rod Photoreceptor Cells/metabolism , Animals , Genome-Wide Association Study/methods , Humans , Male , Mice
13.
Mol Vis ; 15: 1915-28, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19784390

ABSTRACT

PURPOSE: The pig eye is similar to the human eye in terms of anatomy, vasculature, and photoreceptor distribution, and therefore provides an attractive animal model for research into retinal disease. The purpose of this study was to characterize retinal histology in the developing and mature pig retina using antibodies to well established retinal cell markers commonly used in rodents. METHODS: Eyes were enucleated from fetuses in the 9th week of gestation, 1 week old piglets and 6 months old adult animals. Eyeglobes were fixed and cryosectioned. A panel of antibodies to well established retinal markers was employed for immunohistochemistry. Fluorescently labeled secondary antibodies were used for signal detection, and images were acquired by confocal microscopy. Mouse retina at postnatal day (P) 5 was used as a reference for this study to compare progression of histogenesis. Most of the primary antibodies have previously been used on mouse tissue. RESULTS: Most of the studied markers were detected in midgestation pig retina, and the majority had a similar distribution in pig as in P5 mouse retina. However, rhodopsin immunolabeling was detected in pig retina at midgestation but not in P5 mouse retina. Contrary to findings in all rodents, horizontal cells were Islet1-positive and cones were calbindin-immunoreactive in pig retina, as has also been shown for the primate retina. Recoverin and rhodopsin immunolabeling revealed an increase in the length of photoreceptor segments in 6 months, compared to 1 week old animals. CONCLUSIONS: Comparison with the published data on human retina revealed similar marker distribution and histogenesis progression in the pig and human retina, supporting the pig as a valuable animal model for studies on retinal disease and repair. Furthermore, this study provides information about the dynamics of retinal histogenesis in the pig and validates a panel of antibodies that reliably detects developing and mature retinal cell phenotypes in the pig retina.


Subject(s)
Retina/growth & development , Retina/metabolism , Animals , Biomarkers/metabolism , Calbindin 2 , Calbindins , Cryoultramicrotomy , Eye Proteins/metabolism , Glutamate Decarboxylase/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Ki-67 Antigen/metabolism , LIM-Homeodomain Proteins , Mice , Models, Animal , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/cytology , Neuroglia/metabolism , Nuclear Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Protein Kinase C-alpha/metabolism , Repressor Proteins/metabolism , Retina/cytology , Retina/enzymology , S100 Calcium Binding Protein G/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Sus scrofa , Transcription Factors
14.
Mol Cell Biol ; 26(17): 6623-32, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914744

ABSTRACT

Cybr (also known as Cytip, CASP, and PSCDBP) is an interleukin-12-induced gene expressed exclusively in hematopoietic cells and tissues that associates with Arf guanine nucleotide exchange factors known as cytohesins. Cybr levels are dynamically regulated during T-cell development in the thymus and upon activation of peripheral T cells. In addition, Cybr is induced in activated dendritic cells and has been reported to regulate dendritic cell (DC)-T-cell adhesion. Here we report the generation and characterization of Cybr-deficient mice. Despite the selective expression in hematopoietic cells, there was no intrinsic defect in T- or B-cell development or function in Cybr-deficient mice. The adoptive transfer of Cybr-deficient DCs showed that they migrated efficiently and stimulated proliferation and cytokine production by T cells in vivo. However, competitive stem cell repopulation experiments showed a defect in the abilities of Cybr-deficient T cells to develop in the presence of wild-type precursors. These data suggest that Cybr is not absolutely required for hematopoietic cell development or function, but stem cells lacking Cybr are at a developmental disadvantage compared to wild-type cells. Collectively, these data demonstrate that despite its selective expression in hematopoietic cells, the role of Cybr is limited or largely redundant. Previous in vitro studies using overexpression or short interfering RNA inhibition of the levels of Cybr protein appear to have overestimated its immunological role.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Cross-Priming/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Membrane Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cytokines/pharmacology , Dendritic Cells/drug effects , Exons/genetics , Gene Expression Regulation/drug effects , Gene Targeting , Humans , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Lymphocyte Subsets/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Myeloid Cells/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/drug effects
15.
Prog Retin Eye Res ; 26(5): 453-69, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17662637

ABSTRACT

Aqueous humor is actively produced in the ciliary epithelium of the anterior chamber and has important functions for the eye. Under normal physiological conditions, the inflow and outflow of the aqueous humor are tightly regulated, but in the pathologic state this balance is lost. Aqueous outflow involves structures of the anterior chamber and experiences most resistance at the level of the trabecular meshwork (TM) that acts as a filter. The modulation of the TM structure regulates the filter and its mechanism remains poorly understood. Proteomic analyses have identified cochlin, a protein of poorly understood function, in the glaucomatous TM but not in healthy control TM from human cadaver eyes. The presence of cochlin has subsequently been confirmed by Western and immunohistochemical analyses. Functionally, cochlin undergoes multimerization induced by shear stress and other changes in the microenvironment. Cochlin along with mucopolysaccharide deposits has been found in the TM of glaucoma patients and in the inner ear of subjects affected by the hearing disorder DNFA9, a late-onset, progressive disease that also involves alterations in fluid shear regimes. In vitro, cochlin induces aggregation of primary TM cells suggesting a role in cell adhesion, possibly in mechanosensation, and in modulation of the TM filter.


Subject(s)
Glaucoma/metabolism , Ocular Physiological Phenomena , Proteins/physiology , Animals , Aqueous Humor/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Hearing Disorders/metabolism , Humans , Mutation , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Trabecular Meshwork/metabolism
16.
Mol Endocrinol ; 21(12): 3013-27, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17717072

ABSTRACT

Pubertal development is impaired in mice lacking the basic helix-loop-helix transcription factor Nhlh2. The mechanisms underlying changes in reproduction in Nhlh2-deficient mice (Nhlh2(-/-)) are unclear. Here we show that hypothalamic GnRH-1 content is reduced in adult Nhlh2(-/-) mice as is the number of GnRH-1 neurons localized to mid- and caudal hypothalamic regions. This reduction was detected postnatally after normal migration of GnRH-1 neurons within nasal regions had occurred. Phenotype rescue experiments showed that female Nhlh2(-/-) mice were responsive to estrogen treatment. In contrast, puberty could not be primed in female Nhlh2(-/-) mice with a GnRH-1 regimen. The adenohypophysis of Nhlh2(-/-) mice was hypoplastic although it contained a full complement of the five anterior pituitary cell types. GnRH-1 receptors (GnRHRs) were reduced in Nhlh2(-/-) pituitary gonadotropes as compared with wild type. In vitro assays indicated that Nhlh2 expression is regulated in parallel with GnRHR expression. However, direct transcriptional activity of Nhlh2 on the GnRHR promoter was not found. These results indicate that Nhlh2 plays a role in the development and functional maintenance of the hypothalamic-pituitary-gonadal axis at least at two levels: 1) in the hypothalamus by regulating the number and distribution of GnRH-1 neurons and, 2) in the developing and mature adenohypophysis.


Subject(s)
Aging/physiology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypothalamus/physiopathology , Pituitary Diseases/physiopathology , Sexual Maturation , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Movement , Cells, Cultured , Female , Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/pathology , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Phenotype , Pituitary Diseases/genetics , Pituitary Diseases/pathology
17.
Invest Ophthalmol Vis Sci ; 59(12): 4945-4952, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30347088

ABSTRACT

Purpose: Oral nitisinone has been shown to increase fur and ocular pigmentation in a mouse model of oculocutaneous albinism (OCA) due to hypomorphic mutations in tyrosinase (TYR), OCA1B. This study determines if nitisinone can improve ocular and/or fur pigmentation in a mouse model of OCA type 3 (OCA3), caused by mutation of the tyrosinase-related protein 1 (Tyrp1) gene. Methods: Mice homozygous for a null allele in the Tyrp1 gene (C57BL/6J-Tyrp1 b-J/J) were treated with 8 mg/kg nitisinone or vehicle every other day by oral gavage. Changes in fur and ocular melanin pigmentation were monitored. Mature ocular melanosome number and size were quantified in pigmented ocular structures by electron microscopy. Results: C57BL/6J-Tyrp1 b-J/J mice carry a novel c.403T>A; 404delG mutation in Tyrp1, predicted to result in premature truncation of the TYRP1 protein. Nitisinone treatment resulted in an approximately 7-fold increase in plasma tyrosine concentrations without overt toxicity. After 1 month of treatment, no change in the color of fur or pigmented ocular structures was observed. The distribution of melanosome cross-sectional area was unchanged in ocular tissues. There was no significant difference in the number of pigmented melanosomes in the RPE/choroid of nitisinone-treated and control groups. However, there was a significant difference in the number of pigmented melanosomes in the iris. Conclusions: Treatment of a mouse model of OCA3 with oral nitisinone did not have a favorable clinical effect on melanin production and minimally affected the number of pigmented melanosomes in the iris stroma. As such, treatment of OCA3 patients with nitisinone is unlikely to be therapeutic.


Subject(s)
Albinism, Oculocutaneous/drug therapy , Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Nitrobenzoates/therapeutic use , Administration, Oral , Albinism, Oculocutaneous/blood , Albinism, Oculocutaneous/genetics , Albinism, Oculocutaneous/pathology , Animals , Blotting, Western , Disease Models, Animal , Genotyping Techniques , Melanins/metabolism , Melanosomes/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Electron, Transmission , Oxidoreductases/genetics , Real-Time Polymerase Chain Reaction , Treatment Outcome , Tyrosine/blood
18.
Mol Vis ; 13: 1045-57, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17653049

ABSTRACT

PURPOSE: Retinal progenitor cells (RPCs) and retinal stem cells (RSCs) from rodents and humans have been isolated and characterized in vitro. Transplantation experiments have confirmed their potential as tools for cell replacement in retinal degenerative diseases. The pig represents an ideal pre-clinical animal model to study the impact of transplantation because of the similarity of its eye to the human eye. However, little is known about porcine RPCs and RSCs. We aimed to identify and characterize in vitro RPCs and RSCs from porcine ocular tissues. METHODS: Cells from different subregions of embryonic, postnatal and adult porcine eyes were grown in suspension sphere culture in serum-free medium containing basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF). Growth curves and BrdU incorporation assays were performed to establish the proliferative capacity of isolated porcine retina-derived RPCs and ciliary epithelium (CE)-derived RSCs. Self-renewal potential was investigated by subsphere formation assays. Changes in gene expression were assayed by reverse transcription polymerase chain reaction (RT-PCR) at different passages in culture. Finally, differentiation was induced by addition of serum to the cultures and expression of markers for retinal cell types was detected by immunohistochemical staining with specific antibodies. RESULTS: Dissociated cells from embryonic retina and CE at different postnatal ages generated primary nestin- and Pax6-immunoreactive neurosphere colonies in vitro in numbers that decreased with age. Embryonic and postnatal retina-derived RPCs and young CE-derived RSCs displayed self-renewal capacity, generating secondary neurosphere colonies. However, their self-renewal and proliferation capacity gradually decreased and they became more committed to differentiated states with subsequent passages. The expansion capacity of RPCs and RSCs was higher when they were maintained in monolayer culture. Porcine RPCs and RSCs could be induced to differentiate in vitro to express markers of retinal neurons and glia. CONCLUSIONS: Porcine retina and CE contain RPCs and RSCs which are undifferentiated, self-renewing and multipotent and which show characteristics similar to their human counterparts. Therefore, the pig could be a useful source of cells to further investigate the cell biology of RPCs and RSCs and it could be used as a non-primate large animal model for pre-clinical studies on stem cell-based approaches to regenerative medicine in the retina.


Subject(s)
Retina/cytology , Stem Cells , Aging/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell Separation , Cells, Cultured , Ciliary Body/cytology , Embryo, Mammalian , Epithelial Cells/cytology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Multipotent Stem Cells/cytology , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/metabolism , Neurons/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/cytology , Stem Cells/cytology , Stem Cells/metabolism , Swine
19.
Mol Cell Biol ; 22(14): 4977-83, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12077327

ABSTRACT

Nhlh1 is a basic helix-loop-helix transcription factor whose expression is restricted to the nervous system and which may play a role in neuronal differentiation. To directly study Nhlh1 function, we generated null mice. Homozygous mutant mice were predisposed to premature, adult-onset, unexpected death. Electrocardiograms revealed decreased total heart rate variability, stress-induced arrhythmia, and impaired baroreceptor sensitivity. This predisposition to arrhythmia is a likely cause of the observed death in the mutant mice. Heterozygosity for the closely related transcription factor Nhlh2 increased the severity of the Nhlh1-null phenotype. No signs of primary cardiac structural or conduction abnormalities could be detected upon necropsy of the null mice. The pattern of altered heart rhythm observed in basal and experimental conditions (stress and pharmacologically induced) suggests that a deficient parasympathetic tone may contribute to the arrhythmia in the Nhlh1-null mouse. The expression of Nhlh1 in the developing brain stem and in the vagal nuclei in the wild-type mouse further supports this hypothesis. The Nhlh1 mutant mouse may thus provide a model to investigate the contribution of the autonomic nervous system to arrhythmogenesis.


Subject(s)
Arrhythmias, Cardiac/etiology , Autonomic Nervous System Diseases/etiology , DNA-Binding Proteins/deficiency , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/physiopathology , Basic Helix-Loop-Helix Transcription Factors , Bradycardia/physiopathology , Brain Stem/embryology , Brain Stem/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Disease Models, Animal , Diving/physiology , Electrocardiography , Gene Expression Regulation, Developmental , Gene Targeting , Heart Conduction System/physiopathology , Heart Rate/physiology , Heterozygote , Homozygote , In Situ Hybridization , Longevity/genetics , Mice , Mice, Knockout , Phenotype , Pressoreceptors/physiopathology
20.
Cell Rep ; 20(2): 384-396, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28700940

ABSTRACT

Mutations in CEP290, a transition zone protein in primary cilia, cause diverse ciliopathies, including Leber congenital amaurosis (LCA) and Joubert-syndrome and related disorders (JSRD). We examined cilia biogenesis and function in cells derived from CEP290-LCA and CEP290-JSRD patients. CEP290 protein was reduced in LCA fibroblasts with no detectable impact on cilia; however, optic cups derived from induced pluripotent stem cells (iPSCs) of CEP290-LCA patients displayed less developed photoreceptor cilia. Lack of CEP290 in JSRD fibroblasts resulted in abnormal cilia and decreased ciliogenesis. We observed selectively reduced localization of ADCY3 and ARL13B. Notably, Hedgehog signaling was augmented in CEP290-JSRD because of enhanced ciliary transport of Smoothened and GPR161. These results demonstrate a direct correlation between the extent of ciliogenesis defects in fibroblasts and photoreceptors with phenotypic severity in JSRD and LCA, respectively, and strengthen the role of CEP290 as a selective ciliary gatekeeper for transport of signaling molecules in and out of the cilium.


Subject(s)
Antigens, Neoplasm/genetics , Fibroblasts/metabolism , Neoplasm Proteins/genetics , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Alleles , Animals , Antigens, Neoplasm/metabolism , Cell Cycle Proteins , Cilia , Cytoskeletal Proteins , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homozygote , Humans , Mice , Mice, Knockout , Mutation/genetics , Neoplasm Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL