Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
bioRxiv ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38798688

RESUMO

Mutations in the human Ocular albinism type-1 gene OA1 are associated with abnormal retinal pigment epithelium (RPE) melanogenesis and poor binocular vision resulting from misrouting of ipsilateral retinal ganglion cell (iRGC) axons to the brain. We studied the latter using wild-type (WT) and Oa1-/- mouse eyes. At embryonic stages, the WT RPE-specific Oa1 protein signals through cAMP/Epac1-Erk2-CREB. Following CREB phosphorylation, a pCREB gradient extends from the RPE to the differentiating retinal amacrine and RGCs. In contrast to WT, the Oa1-/- RPE and ventral ciliary-margin-zone, a niche for iRGCs, express less pCREB while their retinas have a disrupted pCREB gradient, indicating Oa1's involvement in pCREB maintenance. Oa1-/- retinas also show hyperproliferation, enlarged nuclei, reduced differentiation, and fewer newborn amacrine and RGCs than WT retinas. Our results demonstrate that Oa1's absence leads to reduced binocular vision through a hyperproliferation-associated block in differentiation that impairs neurogenesis. This may affect iRGC axon's routing to the brain.

2.
CRISPR J ; 5(3): 457-471, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35686978

RESUMO

Mutations in the GPR143 gene cause X-linked ocular albinism type 1 (OA1), a disease that severely impairs vision. We recently generated induced pluripotent stem cells (iPSCs) from skin fibroblasts of an OA1 patient carrying a point mutation in intron 7 of GPR143. This mutation activates a new splice site causing the incorporation of a pseudoexon. In this study, we present a high-performance CRISPR-Cas ribonucleoprotein strategy to permanently correct the GPR143 mutation in these patient-derived iPSCs. Interestingly, the two single-guide RNAs available for SpCas9 did not allow the cleavage of the target region. In contrast, the cleavage achieved with the CRISPR-AsCas12a system promoted homology-directed repair at a high rate. The CRISPR-AsCas12a-mediated correction did not alter iPSC pluripotency or genetic stability, nor did it result in off-target events. Moreover, we highlight that the disruption of the pathological splice site caused by CRISPR-AsCas12a-mediated insertions/deletions also rescued the normal splicing of GPR143 and its expression level.


Assuntos
Albinismo Ocular , Células-Tronco Pluripotentes Induzidas , Albinismo Ocular/genética , Albinismo Ocular/metabolismo , Albinismo Ocular/patologia , Sistemas CRISPR-Cas/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Edição de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação
3.
Stem Cell Res ; 33: 274-277, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30513407

RESUMO

Ocular albinism type 1 is a genetic eye disease caused by mutations in the GPR143 gene. Little is known about the molecular pathways involved in this disease and no therapeutic candidate has been identified as yet. Here we report the generation of an iPSC line from the skin fibroblasts of a patient with a mutation in the GPR143 gene using Sendai Virus vectors. This new iPSC line will allow a better understanding of the Ocular Albinism type 1 disease and to screen for potential therapeutic candidates.


Assuntos
Albinismo Ocular/genética , Proteínas do Olho/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteínas de Membrana/genética , Adulto , Humanos , Masculino , Mutação
4.
PLoS One ; 13(3): e0194004, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29538408

RESUMO

Extracellular vesicles (EVs) released by virtually every cell of all organisms are involved in processes of intercellular communication through the delivery of their functional mRNAs, proteins and bioactive lipids. We previously demonstrated that mouse embryonic stem cell-released EVs (mESEVs) are able to transfer their content to different target retinal cells, inducing morphological and biochemical changes in them. The main objective of this paper is to characterize EVs derived from human embryonic stem cells (hESEVs) and investigate the effects that they have on cultured retinal glial, progenitor Müller cells, which are known to give rise to retinal neurons under specific conditions. This would allow us to establish if hESEVs have a pro-regenerative potential not yet described that could be used in the future for treatment of human retinal degenerative diseases. Initially, we showed that hESEVs are heterogeneous in size, contain mRNAs and proteins involved in the induction and maintenance of stem cell pluripotency and can be internalized by cultured Müller cells. After a single exposure to hESEVs these cells display changes in their gene expression profile, and with multiple exposures they de-differentiate and trans-differentiate into retinal neuronal precursors. hESEVs were then fractionated into microvesicles (MVs) and exosomes (EXOs), which were characterized by size, specific surface proteins and biochemical/molecular components. We demonstrate that despite the similar internalization of non-fractionated hESEVs, MVs and EXOs by Müller progenitor cells, in vitro, only the release of MVs' cargo into the cells' cytoplasm induces specific changes in their levels of pluripotency mRNAs and early retinal proteins. EXOs do not produce any detectable effect. Thus, we conclude that MVs and MVs-containing hESEVs are promising agents that possibly could promote the regeneration of diseased or damaged retinas in vivo through inducing glial Müller cells to become replacement neurons.


Assuntos
Células Ependimogliais/fisiologia , Vesículas Extracelulares/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/fisiologia , Células Cultivadas , Células Ependimogliais/metabolismo , Exossomos/metabolismo , Exossomos/fisiologia , Vesículas Extracelulares/metabolismo , Células HEK293 , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Neuroglia/metabolismo , Neuroglia/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , RNA Mensageiro/metabolismo , Regeneração/fisiologia , Retina/metabolismo , Retina/fisiologia , Transcriptoma/fisiologia
5.
PLoS One ; 11(9): e0162273, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27607449

RESUMO

Ocular albinism type 1 (OA), caused by mutations in the OA1 gene, encodes a G-protein coupled receptor, OA1, localized in melanosomal membranes of the retinal pigment epithelium (RPE). This disorder is characterized by both RPE macro-melanosomes and abnormal decussation of ganglion cell axons at the brain's optic chiasm. We demonstrated previously that Oa1 specifically activates Gαi3, which also signals in the Oa1 transduction pathway that regulates melanosomal biogenesis. In this study, we screened the human Gαi3 gene, GNAI3, in DNA samples from 26 patients who had all clinical characteristics of OA but in whom a specific mutation in the OA1 gene had not been found, and in 6 normal control individuals. Using the Agilent HaloPlex Target Enrichment System and next-generation sequencing (NGS) on the Illumina MiSeq platform, we identified 518 variants after rigorous filtering. Many of these variants were corroborated by Sanger sequencing. Overall, 98.8% coverage of the GNAI3 gene was obtained by the HaloPlex amplicons. Of all variants, 6 non-synonymous and 3 synonymous were in exons, 41 in a non-coding exon embedded in the 3' untranslated region (UTR), 6 in the 5' UTR, and 462 in introns. These variants included novel SNVs, insertions, deletions, and a frameshift mutation. All were found in at least one patient but none in control samples. Using computational methods, we modeled the GNAI3 protein and its non-synonymous exonic mutations and determined that several of these may be the cause of disease in the patients studied. Thus, we have identified GNAI3 as a second gene possibly responsible for X-linked ocular albinism.


Assuntos
Albinismo Ocular/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Albinismo Ocular/diagnóstico , Pareamento de Bases/genética , Sequência de Bases , Éxons/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Loci Gênicos , Heterozigoto , Homozigoto , Humanos , Íntrons/genética , Modelos Moleculares , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Reprodutibilidade dos Testes , Análise de Sequência de DNA
6.
Adv Exp Med Biol ; 854: 563-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427460

RESUMO

Mouse embryonic stem cells (mESCs) release into the medium in which they are cultured heterogeneous populations of microvesicles (mESMVs), important components of cell-cell communication, that transfer their contents not only to other stem cells but also to cells of other origins. The purpose of these studies was to demonstrate that ESMVs could be the signals that lead the retinal progenitor Müller cells to de-differentiate and re-entry the cell cycle, followed by differentiation along retinal lineages. Indeed, we found that ESMVs induce these processes and change Müller cells' microenvironment towards a more permissive state for tissue regeneration.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Células-Tronco Embrionárias/citologia , Regeneração/fisiologia , Retina/fisiologia , Animais , Desdiferenciação Celular/genética , Desdiferenciação Celular/fisiologia , Micropartículas Derivadas de Células/genética , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos Transgênicos , Microscopia Confocal , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regeneração/genética , Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
PLoS One ; 8(9): e76240, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098784

RESUMO

PURPOSE: Ocular Albinism type 1 (OA1) is a disease caused by mutations in the OA1 gene and characterized by the presence of macromelanosomes in the retinal pigment epithelium (RPE) as well as abnormal crossing of the optic axons at the optic chiasm. We showed in our previous studies in mice that Oa1 activates specifically Gαi3 in its signaling pathway and thus, hypothesized that a constitutively active Gαi3 in the RPE of Oa1-/- mice might keep on the Oa1 signaling cascade and prevent the formation of macromelanosomes. To test this hypothesis, we have generated transgenic mice that carry the constitutively active Gαi3 (Q204L) protein in the RPE of Oa1-/- mice and are now reporting the effects that the transgene produced on the Oa1-/- RPE phenotype. METHODS: Transgenic mice carrying RPE-specific expression of the constitutively active Gαi3 (Q204L) were generated by injecting fertilized eggs of Oa1-/- females with a lentivirus containing the Gαi3 (Q204L) cDNA. PCR, Southern blots, Western blots and confocal microscopy were used to confirm the presence of the transgene in the RPE of positive transgenic mice. Morphometrical analyses were performed using electron microscopy to compare the size and number of melanosomes per RPE area in putative Oa1-/-, Gαi3 (Q204L) transgenic mice with those of wild-type NCrl and Oa1-/- mice. RESULTS: We found a correlation between the presence of the constitutively active Gαi3 (Q204L) transgene and the rescue of the normal phenotype of RPE melanosomes in Oa1-/-, Gαi3 (Q204L) mice. These mice have higher density of melanosomes per RPE area and a larger number of small melanosomes than Oa1-/- mice, and their RPE phenotype is similar to that of wild-type mice. CONCLUSIONS: Our results show that a constitutively active Gαi3 protein can by-pass the lack of Oa1 protein in Oa1-/- mice and consequently rescue the RPE melanosomal phenotype.


Assuntos
Albinismo Ocular/patologia , Proteínas do Olho/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Glicoproteínas de Membrana/genética , Receptores Acoplados a Proteínas G/genética , Epitélio Pigmentado da Retina/anormalidades , Transdução de Sinais/fisiologia , Animais , Southern Blotting , Western Blotting , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica , Receptores Acoplados a Proteínas G/deficiência , Epitélio Pigmentado da Retina/metabolismo
8.
J Biol Chem ; 288(14): 9742-9754, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23386608

RESUMO

The novel rhomboid-like protein RHBDD2 is distantly related to rhomboid proteins, a group of highly specialized membrane-bound proteases that catalyze regulated intramembrane proteolysis. In retina, RHBDD2 is expressed from embryonic stages to adulthood, and its levels show age-dependent changes. RHBDD2 is distinctly abundant in the perinuclear region of cells, and it localizes to their Golgi. A glycine zipper motif present in one of the transmembrane domains of RHBDD2 is important for its packing into the Golgi membranes. Its deletion causes dislodgment of RHBDD2 from the Golgi. A specific antibody against RHBDD2 recognizes two forms of the protein, one with low (39 kDa; RHBDD2(L)) and the other with high (117 kDa; RHBDD2H) molecular masses in mouse retinal extracts. RHBDD2(L) seems to be ubiquitously expressed in all retinal cells. In contrast, RHBDD2H seems to be present only in the outer segments of cone photoreceptors and may correspond to a homotrimer of RHBDD2(L). This protein consistently co-localizes with S- and M-types of cone opsins. We identified a homozygous mutation in the human RHBDD2 gene, R85H, that co-segregates with disease in affected members of a family with autosomal recessive retinitis pigmentosa. Our findings suggest that the RHBDD2 protein plays important roles in the development and normal function of the retina.


Assuntos
Endopeptidases/biossíntese , Endopeptidases/fisiologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/fisiologia , Retina/metabolismo , Retinose Pigmentar/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Feminino , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Glicina/química , Complexo de Golgi/metabolismo , Células HEK293 , Homozigoto , Humanos , Imuno-Histoquímica/métodos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Opsinas/química , Gravidez , Prenhez , Retina/embriologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Homologia de Sequência de Aminoácidos
9.
PLoS One ; 7(11): e50417, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226281

RESUMO

Cell-derived microvesicles (MVs), recognized as important components of cell-cell communication, contain mRNAs, miRNAs, proteins and lipids and transfer their bioactive contents from parent cells to cells of other origins. We have studied the effect that MVs released from embryonic stem cells (ESMVs) have on retinal progenitor Müller cells. Cultured human Müller cells were exposed to mouse ESMVs every 48 hours for a total of 9 treatments. Morphological changes were observed by light microscopy in the treated cells, which grew as individual heterogeneous cells, compared to the uniform, spindle-like adherent cellular sheets of untreated cells. ESMVs transferred to Müller cells embryonic stem cell (ESC) mRNAs involved in the maintenance of pluripotency, including Oct4 and Sox2, and the miRNAs of the 290 cluster, important regulators of the ESC-specific cell cycle. Moreover, ESMV exposure induced up-regulation of the basal levels of endogenous human Oct4 mRNA in Müller cells. mRNA and miRNA microarrays of ESMV-treated vs. untreated Müller cells revealed the up-regulation of genes and miRNAs involved in the induction of pluripotency, cellular proliferation, early ocular genes and genes important for retinal protection and remodeling, as well as the down-regulation of inhibitory and scar-related genes and miRNAs involved in differentiation and cell cycle arrest. To further characterize the heterogeneous cell population of ESMV-treated Müller cells, their expression of retinal cell markers was compared to that in untreated control cells by immunocytochemistry. Markers for amacrine, ganglion and rod photoreceptors were present in treated but not in control Müller cells. Together, our findings indicate that ESMs induce de-differentiation and pluripotency in their target Müller cells, which may turn on an early retinogenic program of differentiation.


Assuntos
Vesículas Citoplasmáticas/química , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/genética , Retina/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Forma Celular , Vesículas Citoplasmáticas/metabolismo , Células-Tronco Embrionárias/citologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Células Fotorreceptoras de Vertebrados/metabolismo , Células-Tronco Pluripotentes/citologia , RNA Mensageiro/metabolismo , Retina/citologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais
10.
PLoS One ; 7(5): e35317, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693546

RESUMO

Zbed4, a member of the BED subclass of Zinc-finger proteins, is expressed in cone photoreceptors and glial Müller cells of human retina whereas it is only present in Müller cells of mouse retina. To characterize structural and functional properties of Zbed4, enough amounts of purified protein were needed. Thus, recombinant Zbed4 was expressed in E. coli and its refolding conditions optimized for the production of homogenous and functionally active protein. Zbed4's secondary structure, determined by circular dichroism spectroscopy, showed that this protein contains 32% α-helices, 18% ß-sheets, 20% turns and 30% unordered structures. CASTing was used to identify the target sites of Zbed4 in DNA. The majority of the DNA fragments obtained contained poly-Gs and some of them had, in addition, the core signature of GC boxes; a few clones had only GC-boxes. With electrophoretic mobility shift assays we demonstrated that Zbed4 binds both not only to DNA and but also to RNA oligonucleotides with very high affinity, interacting with poly-G tracts that have a minimum of 5 Gs; its binding to and GC-box consensus sequences. However, the latter binding depends on the GC-box flanking nucleotides. We also found that Zbed4 interacts in Y79 retinoblastoma cells with nuclear and cytoplasmic proteins Scaffold Attachment Factor B1 (SAFB1), estrogen receptor alpha (ERα), and cellular myosin 9 (MYH9), as shown with immunoprecipitation and mass spectrometry studies as well as gel overlay assays. In addition, immunostaining corroborated the co-localization of Zbed4 with these proteins. Most importantly, in vitro experiments using constructs containing promoters of genes directing expression of the luciferase gene, showed that Zbed4 transactivates the transcription of those promoters with poly-G tracts.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/genética , DNA/metabolismo , Proteínas Recombinantes/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Citoplasma/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Redobramento de Proteína , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Retina/metabolismo , Especificidade por Substrato , Transativadores/química , Transativadores/genética , Fatores de Transcrição
11.
PLoS One ; 6(9): e24376, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931697

RESUMO

BACKGROUND: Ocular albinism type 1, an X-linked disease characterized by the presence of enlarged melanosomes in the retinal pigment epithelium (RPE) and abnormal crossing of axons at the optic chiasm, is caused by mutations in the OA1 gene. The protein product of this gene is a G-protein-coupled receptor (GPCR) localized in RPE melanosomes. The Oa1-/- mouse model of ocular albinism reproduces the human disease. Oa1 has been shown to immunoprecipitate with the Gαi subunit of heterotrimeric G proteins from human skin melanocytes. However, the Gαi subfamily has three highly homologous members, Gαi1, Gαi2 and Gαi3 and it is possible that one or more of them partners with Oa1. We had previously shown by in-vivo studies that Gαi3-/- and Oa1-/- mice have similar RPE phenotype and decussation patterns. In this paper we analyze the specificity of the Oa1-Gαi interaction. METHODOLOGY: By using the genetic mouse models Gαi1-/-, Gαi2-/-, Gαi3-/- and the double knockout Gαi1-/-, Gαi3-/- that lack functional Gαi1, Gαi2, Gαi3, or both Gαi1 and Gαi3 proteins, respectively, we show that Gαi3 is critical for the maintenance of a normal melanosomal phenotype and that its absence is associated with changes in melanosomal size and density. GST-pull-down and immunoprecipitation assays conclusively demonstrate that Gαi3 is the only Gαi that binds to Oa1. Western blots show that Gαi3 expression is barely detectable in the Oa1-/- RPE, strongly supporting a previously unsuspected role for Gαi3 in melanosomal biogenesis. CONCLUSION: Our results identify the Oa1 transducer Gαi3 as the first downstream component in the Oa1 signaling pathway.


Assuntos
Proteínas do Olho/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Melanossomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Tamanho das Organelas , Receptores Acoplados a Proteínas G/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Adenosina Difosfato Ribose/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Cromatografia Líquida , Eletrorretinografia , Proteínas do Olho/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Humanos , Espectrometria de Massas , Melanossomas/ultraestrutura , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Dados de Sequência Molecular , Forma das Organelas , Ligação Proteica , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/deficiência , Epitélio Pigmentado da Retina/fisiologia , Epitélio Pigmentado da Retina/ultraestrutura
12.
Mol Vis ; 17: 2011-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21850176

RESUMO

PURPOSE: ZBED4, a protein in cones and Müller cells of human retina, may play important functions as a transcriptional activator of genes expressed in those cells or as a co-activator/repressor of their nuclear hormone receptors. To begin investigating these potential roles of ZBED4, we studied the developmental expression and localization of both the Zbed4 mRNA and protein of mouse retina. METHODS: northern blots showed the presence of Zbed4 mRNA in retina and other mouse tissues, and western blots showed the nuclear and cytoplasmic expression of Zbed4 at different developmental times. Antibodies against Zbed4 and specific retinal cell markers were used for retinal immunohistochemistry. RESULTS: Zbed4 mRNA was present at different levels in all the mouse tissues analyzed. The Zbed4 protein was barely detectable at embryonic day (E)14.5 but was clearly seen at E16 at both retinal outer and vitreal borders and throughout the retina by E18 and postnatal day 0 (P0). Thereafter, Zbed4 expression was more restricted to the inner retina. While ZBED4 is localized in cones and endfeet of Müller cells of human retina, in adult mouse retina Zbed4 is only detected in Müller cell endfeet and processes. The same localization of Zbed4 was observed in rat retina. In early development, Zbed4 is mainly present in the nuclear fraction of the mouse retina, and in adulthood it becomes more enriched in the cytoplasmic fraction. CONCLUSIONS: The patterns of spatial and temporal expression of Zbed4 in the mouse retina suggest a possible involvement of this protein in retinal morphogenesis and Müller cell function.


Assuntos
Animais Recém-Nascidos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Morfogênese/genética , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos/genética , Northern Blotting , Western Blotting , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Retina/citologia , Retina/embriologia , Células Fotorreceptoras Retinianas Cones/citologia , Fatores de Transcrição/genética
13.
PLoS One ; 6(6): e20707, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738583

RESUMO

Retinoschisin is encoded by the gene responsible for X-linked retinoschisis (XLRS), an early onset macular degeneration that results in a splitting of the inner layers of the retina and severe loss in vision. Retinoschisin is predominantly expressed and secreted from photoreceptor cells as a homo-oligomer protein; it then associates with the surface of retinal cells and maintains the retina cellular architecture. Many missense mutations in the XLRS1 gene are known to cause intracellular retention of retinoschisin, indicating that the secretion process of the protein is a critical step for its normal function in the retina. However, the molecular mechanisms underlying retinoschisin's secretion remain to be fully elucidated. In this study, we investigated the role of the F-actin cytoskeleton in the secretion of retinoschisin by treating Weri-Rb1 cells, which are known to secrete retinoschisin, with cytochalasin D, jasplakinolide, Y-27632, and dibutyryl cGMP. Our results show that cytochalasin D and jasplakinolide inhibit retinoschisin secretion, whereas Y-27632 and dibutyryl cGMP enhance secretion causing F-actin alterations. We also demonstrate that high concentrations of taxol, which hyperpolymerizes microtubules, inhibit retinoschisin secretion. Our data suggest that retinoschisin secretion is regulated by the F-actin cytoskeleton, that cGMP or inhibition of ROCK alters F-actin structure enhancing the secretion, and that the microtubule cytoskeleton is also involved in this process.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas do Olho/metabolismo , Microtúbulos/metabolismo , Amidas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Depsipeptídeos/farmacologia , Dibutiril GMP Cíclico/farmacologia , Humanos , Imuno-Histoquímica , Microtúbulos/efeitos dos fármacos , Paclitaxel/farmacologia , Piridinas/farmacologia , Retinosquise/metabolismo , Moduladores de Tubulina/farmacologia
14.
Adv Exp Med Biol ; 664: 79-87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20238005

RESUMO

To identify genes expressed in cone photoreceptors, we previously carried out subtractive hybridization and microarrays of retinal mRNAs from normal and cd (cone degeneration) dogs. One of the isolated genes encoded ZBED4, a novel protein that in human retina is localized to cone photoreceptors and glial Müller cells. ZBED4 is distributed between nuclear and cytoplasmic fractions of the retina and it readily forms homodimers, probably as a consequence of its hATC dimerization domain. In addition, the ZBED4 sequence has several domains that suggest it may function as part of a co-activator complex facilitating the activation of nuclear receptors and other factors (BED finger domains) or as a co-activator/co-repressor of nuclear hormone receptors (LXXLL motifs). We have identified several putative ZBED4-interacting proteins and one of them is precisely a co-repressor of the estrogen receptor alpha.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho/metabolismo , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas do Olho/química , Proteínas do Olho/genética , Citometria de Fluxo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Espectrometria de Massas , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Frações Subcelulares/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transfecção
15.
Am J Hum Genet ; 84(5): 683-91, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19409519

RESUMO

Cone-rod dystrophy (CRD) is an inherited progressive retinal dystrophy affecting the function of cone and rod photoreceptors. By autozygosity mapping, we identified null mutations in the ADAM metallopeptidase domain 9 (ADAM9) gene in four consanguineous families with recessively inherited early-onset CRD. We also found reduced photoreceptor responses in Adam9 knockout mice, previously reported to be asymptomatic. In 12-month-old knockout mice, photoreceptors appear normal, but the apical processes of the retinal pigment epithelium (RPE) cells are disorganized and contact between photoreceptor outer segments (POSs) and the RPE apical surface is compromised. In 20-month-old mice, there is clear evidence of progressive retinal degeneration with disorganized POS and thinning of the outer nuclear layer (ONL) in addition to the anomaly at the POS-RPE junction. RPE basal deposits and macrophages were also apparent in older mice. These findings therefore not only identify ADAM9 as a CRD gene but also identify a form of pathology wherein retinal disease first manifests at the POS-RPE junction.


Assuntos
Proteínas ADAM/genética , Proteínas de Membrana/genética , Degeneração Retiniana/genética , Retinose Pigmentar/genética , Animais , Consanguinidade , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Knockout , Mutação , Linhagem , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/patologia
16.
Invest Ophthalmol Vis Sci ; 50(8): 3580-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19369242

RESUMO

PURPOSE: To characterize the ZBED4 cDNA identified by subtractive hybridization and microarray of retinal cone degeneration (cd) adult dog mRNA from mRNA of normal dog retina. METHODS: The cDNA library obtained from subtractive hybridization was arrayed and screened with labeled amplicons from normal and cd dog retinas. Northern blot analysis was used to verify ZBED4 mRNA expression in human retina. Flow cytometry sorted peanut agglutinin (PNA)-labeled cones from dissociated mouse retinas, and quantitative RT-PCR (QPCR) was used to measure ZBED4 mRNA levels in these cone cells. Immunohistochemistry localized ZBED4 in human retinas. Expression of ZBED4 mRNA transiently transfected into HEK293 cells was analyzed by immunofluorescence. ZBED4 subcellular localization was determined with Western blot analysis. RESULTS: One of 80 cDNAs differentially expressed in normal and cd dog retinas corresponded to a novel gene, ZBED4, which is also expressed in human and mouse retinas. ZBED4 mRNA was found to be present in cone photoreceptors. When ZBED4 cDNA was transfected into HEK293 cells, the expressed protein showed nuclear localization. However, in human retinas, ZBED4 was localized to cone nuclei, inner segments, and pedicles, as well as to Müller cell endfeet. Confirming these immunohistochemical results, the 135-kDa ZBED4 was found in both the nuclear and cytosolic extracts of human retinas. ZBED4 has four predicted DNA-binding domains, a dimerization domain, and two LXXLL motifs characteristic of coactivators/corepressors of nuclear hormone receptors. CONCLUSIONS: ZBED4 cellular/subcellular localization and domains suggest a regulatory role for this protein, which may exert its effects in cones and Müller cells through multiple ways of action.


Assuntos
Proteínas de Ligação a DNA/genética , Doenças do Cão/genética , Regulação da Expressão Gênica/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/veterinária , Transativadores/genética , Fatores de Transcrição/genética , Animais , Northern Blotting , Células Cultivadas , DNA Complementar/isolamento & purificação , Doenças do Cão/patologia , Cães , Citometria de Fluxo , Humanos , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Análise em Microsséries , Neuroglia/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Coelhos , Células Fotorreceptoras Retinianas Cones/patologia , Degeneração Retiniana/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Dedos de Zinco
17.
PLoS One ; 4(3): e4722, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19266099

RESUMO

Microvesicles are plasma membrane-derived vesicles released into the extracellular environment by a variety of cell types. Originally characterized from platelets, microvesicles are a normal constituent of human plasma, where they play an important role in maintaining hematostasis. Microvesicles have been shown to transfer proteins and RNA from cell to cell and they are also believed to play a role in intercellular communication. We characterized the RNA and protein content of embryonic stem cell microvesicles and show that they can be engineered to carry exogenously expressed mRNA and protein such as green fluorescent protein (GFP). We demonstrate that these engineered microvesicles dock and fuse with other embryonic stem cells, transferring their GFP. Additionally, we show that embryonic stem cells microvesicles contain abundant microRNA and that they can transfer a subset of microRNAs to mouse embryonic fibroblasts in vitro. Since microRNAs are short (21-24 nt), naturally occurring RNAs that regulate protein translation, our findings open up the intriguing possibility that stem cells can alter the expression of genes in neighboring cells by transferring microRNAs contained in microvesicles. Embryonic stem cell microvesicles may be useful therapeutic tools for transferring mRNA, microRNAs, protein, and siRNA to cells and may be important mediators of signaling within stem cell niches.


Assuntos
Micropartículas Derivadas de Células/genética , Células-Tronco Embrionárias/ultraestrutura , MicroRNAs/metabolismo , Animais , Transporte Biológico/genética , Comunicação Celular/genética , Micropartículas Derivadas de Células/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Fluorescência Verde , Camundongos , MicroRNAs/análise
18.
Invest Ophthalmol Vis Sci ; 50(6): 2591-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19218616

RESUMO

PURPOSE: Rod cGMP-phosphodiesterase, a key enzyme in visual transduction, is important for retinal integrity and function. Mutations in the gene encoding the phosphodiesterase beta-subunit (PDEbeta) cause retinal degeneration in animals and humans. Here the authors tested the hypothesis that elements in the 3' untranslated region (3' UTR) of the PDEbeta gene are involved in the regulation of PDEbeta expression. METHODS: Involvement of the 3' UTR of PDEbeta mRNA in the regulation of PDEbeta expression was assessed by Y-79 retinoblastoma cells or the heads of Xenopus laevis tadpoles with constructs containing the SV40 or PDEbeta promoter, the luciferase cDNA, and either the SV40 or the PDEbeta 3' UTR (or fragments of its sequence). RESULTS: Compared with the SV40 3' UTR (used as control), the entire PDEbeta 3' UTR decreased reporter gene expression in Y-79 retinoblastoma cells as well as in SY5Y neuroblastoma and 293 human embryonic kidney cell lines. However, the authors observed that two 100-nucleotide fragments from the PDEbeta 3' UTR increased while its noncanonical poly-adenylation signal abolished reporter gene expression in Y-79 retinoblastoma cells and in ex vivo experiments using Xenopus tadpole heads. In particular, an 11-nucleotide element (EURE) in one of the 100-nucleotide fragments was responsible for the upregulation of luciferase expression. CONCLUSIONS: These studies indicate that the 3' UTR of the PDEbeta mRNA is involved in the complex regulation of this gene's expression in the retina. Moreover, the results show that the PDEbeta poly-A signal has a dominant inhibitory effect over two other regions in the 3' UTR that stimulate gene expression.


Assuntos
Regiões 3' não Traduzidas/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , RNA Mensageiro/genética , Sequências Reguladoras de Ácido Nucleico , Animais , Sequência de Bases , Embrião não Mamífero/metabolismo , Genes Reporter , Humanos , Rim/embriologia , Rim/metabolismo , Dados de Sequência Molecular , Neuroblastoma/genética , Retina/metabolismo , Neoplasias da Retina/genética , Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas/metabolismo , Regulação para Cima , Xenopus laevis/genética
19.
Exp Eye Res ; 87(3): 168-75, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18653181

RESUMO

Despite extensive research on many of the genes responsible for inherited retinal degenerations leading to blindness, no effective treatment is currently available for patients affected with these diseases. Among the therapeutic approaches tested on animal models of human retinal degeneration, gene therapy using different types of viral vectors as delivery agents has yielded promising results. We report here our results on a non-invasive, non-viral delivery approach using transscleral iontophoresis for transfer of plasmid DNA into mouse retina. Proof of principle experiments were carried out using plasmid containing GFP cDNA to demonstrate expression of the transferred gene in the retina after single applications of iontophoresis. Various parameters for multiple applications of iontophoresis were optimized to sustain GFP gene expression in mouse photoreceptors. Subsequently, repeated iontophoresis of plasmid containing normal cGMP-phosphodiesterase beta-subunit (beta-PDE) cDNA was performed in the rd1 mouse, an animal model of autosomal recessive retinitis pigmentosa caused by a mutant beta-PDE gene. In normal mice, transscleral iontophoresis of the GFP plasmid provided a significant increase in fluorescence of the retina in the treated versus non-treated eyes. In rd1 mice, repeated iontophoresis of beta-PDE cDNA plasmid partially rescued photoreceptors morphologically, as observed by microscopy, and functionally, as recorded on ERG measurements, without adverse effects. Therefore, transscleral iontophoresis of plasmid DNA containing therapeutic genes may be an efficient, safe and non-invasive method for the treatment of retinal degenerations.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Iontoforese/métodos , Retinose Pigmentar/terapia , Animais , Western Blotting , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , DNA Complementar/genética , Modelos Animais de Doenças , Eletrorretinografia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Confocal , Plasmídeos , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia
20.
Invest Ophthalmol Vis Sci ; 49(7): 3245-52, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18378571

RESUMO

PURPOSE: Ocular albinism type 1 (OA1) is characterized by abnormalities in retinal pigment epithelium (RPE) melanosomes and misrouting of optic axons. The OA1 gene encodes a G-protein-coupled receptor (GPCR) that coimmunoprecipitates with the G alpha i-subunit of heterotrimeric G-proteins from human melanocyte extracts. This study was undertaken to test whether one of the G alpha i proteins, G alpha i3, signals in the same pathway as OA1 to regulate melanosome biogenesis and axonal growth through the optic chiasm. METHODS: Adult G alpha i3(-/-) and Oa1(-/-) mice were compared with their respective control mice (129Sv and B6/NCrl) to study the effects of the loss of G alpha i3 or Oa1 function. Light and electron microscopy were used to analyze the morphology of the retina and the size and density of RPE melanosomes, electroretinograms to study retinal function, and retrograde labeling to investigate the size of the uncrossed optic pathway. RESULTS: Although G alpha i3(-/-) and Oa1(-/-) photoreceptors were comparable to those of the corresponding control retinas, the density of their RPE melanosomes was significantly lower than in control RPEs. In addition, the RPE cells of G alpha i3(-/-) and Oa1(-/-) mice showed abnormal melanosomes that were far larger than the largest 129Sv and B6/NCrl melanosomes, respectively. Although G alpha i3(-/-) and Oa1(-/-) mice had normal results on electroretinography, retrograde labeling showed a significant reduction from control in the size of their ipsilateral retinofugal projections. CONCLUSIONS: These results indicate that G alpha i3, like Oa1, plays an important role in melanosome biogenesis. Furthermore, they suggest a common Oa1-G alpha i3 signaling pathway that ultimately affects axonal growth through the optic chiasm.


Assuntos
Proteínas do Olho/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Melanossomas/fisiologia , Glicoproteínas de Membrana/metabolismo , Quiasma Óptico/fisiologia , Epitélio Pigmentado Ocular/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Axônios/fisiologia , Proteínas de Transporte/metabolismo , Contagem de Células , Eletrorretinografia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Membranas Intracelulares/metabolismo , Melanossomas/ultraestrutura , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Fenótipo , Epitélio Pigmentado Ocular/ultraestrutura , Receptores Acoplados a Proteínas G/deficiência , Retina/citologia , Retina/fisiologia , Células Ganglionares da Retina/citologia , Transdução de Sinais/fisiologia , Transmissão Sináptica , Vias Visuais/citologia , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...