Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 33(6): 491-500, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-37971355

RESUMO

Pathogenic variants in the highly conserved OVOL2 promoter region cause posterior polymorphous corneal dystrophy (PPCD) 1 by inducing an ectopic expression of the endothelial OVOL2 mRNA. Here we produced an allelic series of Ovol2 promoter mutations in the mouse model including the heterozygous c.-307T>C variant (RefSeq NM_021220.4) causing PPCD1 in humans. Despite the high evolutionary conservation of the Ovol2 promoter, only some alterations of its sequence had phenotypic consequences in mice. Four independent sequence variants in the distal part of the Ovol2 promoter had no significant effect on endothelial Ovol2 mRNA level or caused any ocular phenotype. In contrast, the mutation c.-307T>C resulted in increased Ovol2 expression in the corneal endothelium. However, only a small fraction of adult mice c.-307T>C heterozygotes developed ocular phenotypes such as irido-corneal adhesions, and corneal opacity. Interestingly, phenotypic penetrance was increased at embryonic stages. Notably, c.-307T>C mutation is located next to the Ovol1/Ovol2 transcription factor binding site. Mice carrying an allele with a deletion encompassing the Ovol2 binding site c.-307_-320del showed significant Ovol2 gene upregulation in the cornea endothelium and exhibited phenotypes similar to the c.-307T>C mutation. In conclusion, although the mutations c.-307T>C and -307_-320del lead to a comparably strong increase in endothelial Ovol2 expression as seen in PPCD1 patients, endothelial dystrophy was not observed in the mouse model, implicating species-specific differences in endothelial cell biology. Nonetheless, the emergence of dominant ocular phenotypes associated with Ovol2 promoter variants in mice implies a potential role of this gene in eye development and disease.


Assuntos
Distrofias Hereditárias da Córnea , Adulto , Humanos , Animais , Camundongos , Fenótipo , Distrofias Hereditárias da Córnea/genética , Endotélio Corneano , Modelos Animais de Doenças , RNA Mensageiro , Fatores de Transcrição/genética
2.
Dev Biol ; 491: 1-12, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36049534

RESUMO

Mammalian corneal development is a multistep process, including formation of the corneal epithelium (CE), endothelium and stroma during embryogenesis, followed by postnatal stratification of the epithelial layers and continuous renewal of the epithelium to replace the outermost corneal cells. Here, we employed the Cre-loxP system to conditionally deplete Pax6 proteins in two domains of ocular cells, i.e., the ocular surface epithelium (cornea, limbus and conjunctiva) (OSE) or postnatal CE via K14-cre or Aldh3-cre, respectively. Earlier and broader inactivation of Pax6 in the OSE resulted in thickened OSE with CE and limbal cells adopting the conjunctival keratin expression pattern. More restricted depletion of Pax6 in postnatal CE resulted in an abnormal cornea marked by reduced epithelial thickness despite increased epithelial cell proliferation. Immunofluorescence studies revealed loss of intermediate filament Cytokeratin 12 and diffused expression of adherens junction components, together with reduced tight junction protein, Zonula occludens-1. Furthermore, the expression of Cytokeratin 14, a basal cell marker in apical layers, indicates impaired differentiation of CE cells. Collectively, our data demonstrate that Pax6 is essential for maintaining proper differentiation and strong intercellular adhesion in postnatal CE cells, whereas limbal Pax6 is required to prevent the outgrowth of conjunctival cells to the cornea.


Assuntos
Córnea , Epitélio Corneano , Animais , Córnea/metabolismo , Epitélio Corneano/metabolismo , Queratina-12/metabolismo , Queratina-14/metabolismo , Queratinas/metabolismo , Mamíferos/metabolismo , Proteínas de Junções Íntimas/metabolismo
3.
Sci Rep ; 10(1): 9083, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32493941

RESUMO

Conditional gene targeting in mice by means of Cre-loxP strategy represents a powerful approach to study mammalian gene function. This approach is however dependent on the availability of suitable strains of mice with a tissue or time restricted activity of the Cre recombinase. Here we describe Aldh3-Cre transgenic mice as a useful tool to conditionally delete genes in cornea, a specialized transparent tissue found on the anterior-most part of the eye, which acts as a protective barrier and contributes to the refractive power. Using a set of floxed alleles we demonstrate high Aldh3-Cre activity in corneal epithelial cells, corneal stroma and conjunctival epithelial cells at postnatal stages. Aldh3-Cre will thus be particularly beneficial for functional analysis of genes which are vital for postnatal development of cornea and conjunctiva.


Assuntos
Aldeído Desidrogenase/genética , Córnea/fisiologia , Integrases/genética , Camundongos Transgênicos/genética , Camundongos Transgênicos/fisiologia , Alelos , Animais , Túnica Conjuntiva/fisiologia , Células Epiteliais/fisiologia , Deleção de Genes , Marcação de Genes/métodos , Camundongos
4.
Dev Biol ; 433(1): 47-60, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29137925

RESUMO

Polycomb repressive complexes maintain transcriptional repression of genes encoding crucial developmental regulators through chromatin modification. Here we investigated the role of Polycomb repressive complex 2 (PRC2) in retinal development by inactivating its key components Eed and Ezh2. Conditional deletion of Ezh2 resulted in a partial loss of PRC2 function and accelerated differentiation of Müller glial cells. In contrast, inactivation of Eed led to the ablation of PRC2 function at early postnatal stage. Cell proliferation was reduced and retinal progenitor cells were significantly decreased in this mutant, which subsequently caused depletion of Müller glia, bipolar, and rod photoreceptor cells, primarily generated from postnatal retinal progenitor cells. Interestingly, the proportion of amacrine cells was dramatically increased at postnatal stages in the Eed-deficient retina. In accordance, multiple transcription factors controlling amacrine cell differentiation were upregulated. Furthermore, ChIP-seq analysis showed that these deregulated genes contained bivalent chromatin (H3K27me3+ H3K4me3+). Our results suggest that PRC2 is required for proliferation in order to maintain the retinal progenitor cells at postnatal stages and for retinal differentiation by controlling amacrine cell generation.


Assuntos
Complexo Repressor Polycomb 2/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Cromatina/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/metabolismo , Metilação , Camundongos , Neurogênese , Neuroglia/metabolismo , Retina/metabolismo , Retina/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
PLoS Genet ; 12(12): e1006441, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27918583

RESUMO

Lens induction is a classical developmental model allowing investigation of cell specification, spatiotemporal control of gene expression, as well as how transcription factors are integrated into highly complex gene regulatory networks (GRNs). Pax6 represents a key node in the gene regulatory network governing mammalian lens induction. Meis1 and Meis2 homeoproteins are considered as essential upstream regulators of Pax6 during lens morphogenesis based on their interaction with the ectoderm enhancer (EE) located upstream of Pax6 transcription start site. Despite this generally accepted regulatory pathway, Meis1-, Meis2- and EE-deficient mice have surprisingly mild eye phenotypes at placodal stage of lens development. Here, we show that simultaneous deletion of Meis1 and Meis2 in presumptive lens ectoderm results in arrested lens development in the pre-placodal stage, and neither lens placode nor lens is formed. We found that in the presumptive lens ectoderm of Meis1/Meis2 deficient embryos Pax6 expression is absent. We demonstrate using chromatin immunoprecipitation (ChIP) that in addition to EE, Meis homeoproteins bind to a remote, ultraconserved SIMO enhancer of Pax6. We further show, using in vivo gene reporter analyses, that the lens-specific activity of SIMO enhancer is dependent on the presence of three Meis binding sites, phylogenetically conserved from man to zebrafish. Genetic ablation of EE and SIMO enhancers demostrates their requirement for lens induction and uncovers an apparent redundancy at early stages of lens development. These findings identify a genetic requirement for Meis1 and Meis2 during the early steps of mammalian eye development. Moreover, they reveal an apparent robustness in the gene regulatory mechanism whereby two independent "shadow enhancers" maintain critical levels of a dosage-sensitive gene, Pax6, during lens induction.


Assuntos
Olho/crescimento & desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Cristalino/crescimento & desenvolvimento , Proteínas de Neoplasias/genética , Fator de Transcrição PAX6/genética , Animais , Sítios de Ligação , Ectoderma/crescimento & desenvolvimento , Ectoderma/patologia , Elementos Facilitadores Genéticos/genética , Olho/metabolismo , Olho/patologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Humanos , Cristalino/metabolismo , Cristalino/patologia , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Fator de Transcrição PAX6/metabolismo , Peixe-Zebra/genética
6.
PLoS One ; 8(10): e78279, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205179

RESUMO

The Wnt/ß-catenin signaling pathway controls many processes during development, including cell proliferation, cell differentiation and tissue homeostasis, and its aberrant regulation has been linked to various pathologies. In this study we investigated the effect of ectopic activation of Wnt/ß-catenin signaling during lens fiber cell differentiation. To activate Wnt/ß-catenin signaling in lens fiber cells, the transgenic mouse referred to as αA-CLEF was generated, in which the transactivation domain of ß-catenin was fused to the DNA-binding protein LEF1, and expression of the transgene was controlled by αA-crystallin promoter. Constitutive activation of Wnt/ß-catenin signaling in lens fiber cells of αA-CLEF mice resulted in abnormal and delayed fiber cell differentiation. Moreover, adult αA-CLEF mice developed cataract, microphthalmia and manifested downregulated levels of γ-crystallins in lenses. We provide evidence of aberrant expression of cell cycle regulators in embryonic lenses of αA-CLEF transgenic mice resulting in the delay in cell cycle exit and in the shift of fiber cell differentiation to the central fiber cell compartment. Our results indicate that precise regulation of the Wnt/ß-catenin signaling activity during later stages of lens development is essential for proper lens fiber cell differentiation and lens transparency.


Assuntos
Catarata/genética , Diferenciação Celular/genética , Via de Sinalização Wnt/genética , beta Catenina/genética , Animais , Catarata/metabolismo , Ciclo Celular/genética , Cristalinas/genética , Cristalinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cristalino/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Microftalmia/genética , Microftalmia/metabolismo , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , beta Catenina/metabolismo
7.
PLoS One ; 8(5): e63029, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667567

RESUMO

During mouse eye development, all retinal cell types are generated from the population of retina-committed progenitors originating from the neuroepithelium of the optic vesicle. Conditional gene inactivation provides an efficient tool for studying the genetic basis of the developing retina; however, the number of retina-specific Cre lines is limited. Here we report generation of the mRx-Cre BAC transgenic mouse line in which the expression of Cre recombinase is controlled by regulatory sequences of the mouse Rx gene, one of the earliest determinants of retinal development. When mRx-Cre transgenic mice were crossbred with the ROSA26R or ROSA26R-EYFP reporter lines, the Cre activity was observed in the optic sulcus from embryonic day 8.5 onwards and later in all progenitors residing in the neuroepithelium of the optic cup. Our results suggest that mRx-Cre provides a unique tool for functional genetic studies in very early stages of retinal development. Moreover, since eye organogenesis is dependent on the inductive signals between the optic vesicle and head surface ectoderm, the inductive ability of the optic vesicle can be analyzed using mRx-Cre transgenic mice.


Assuntos
Proteínas do Olho/genética , Deleção de Genes , Engenharia Genética/métodos , Proteínas de Homeodomínio/genética , Integrases/metabolismo , Retina/citologia , Células-Tronco/metabolismo , Animais , Feminino , Camundongos , Camundongos Transgênicos , Gravidez , Recombinação Genética , Células-Tronco/citologia , Fatores de Tempo
8.
Genesis ; 48(2): 86-95, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20027618

RESUMO

Lens formation in mouse is critically dependent on proper development of the retinal neuroectoderm that is located close beneath the head surface ectoderm. Signaling from the prospective retina triggers lens-specific gene expression in the surface-ectoderm. Supression of canonical Wnt/beta-catenin signaling in the surface ectoderm is one of the prerequisites for lens development because, as we show here, ectopic Wnt activation in the retina and lens abrogates lens formation. Wnt inhibiton is mediated by signals coming from the retina but its exact mechanism is unknown. We show that Pax6 directly controls expression of several Wnt inhibitors such as Sfrp1, Sfrp2, and Dkk1 in the presumptive lens. In accordance, absence of Pax6 function leads to aberrant canonical Wnt activity in the presumptive lens that subsequently impairs lens development. Thus Pax6 is required for down-regulation of canonical Wnt signaling in the presumptive lens ectoderm.


Assuntos
Ectoderma/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Cristalino/metabolismo , Morfogênese/genética , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , beta Catenina/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Cristalino/embriologia , Camundongos , Camundongos Transgênicos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Retina/metabolismo , Transdução de Sinais/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
9.
Genesis ; 45(4): 157-68, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17410548

RESUMO

beta-Catenin plays a key role in cadherin-mediated cell adhesion as well as in canonical Wnt signaling. To study the role of beta-catenin during eye development, we used conditional Cre/loxP system in mouse to inactivate beta-catenin in developing lens and retina. Inactivation of beta-catenin does not suppress lens fate, but instead results in abnormal morphogenesis of the lens. Using BAT-gal reporter mice, we show that beta-catenin-mediated Wnt signaling is notably absent from lens and neuroretina throughout eye development. The observed defect is therefore likely due to the cytoskeletal role of beta-catenin, and is accompanied by impaired epithelial cell adhesion. In contrast, inactivation of beta-catenin in the nasal ectoderm, an area with active Wnt signaling, results in formation of crystallin-positive ectopic lentoid bodies. These data suggest that, outside of the normal lens, beta-catenin functions as a coactivator of canonical Wnt signaling to suppress lens fate.


Assuntos
Coristoma/genética , Cristalino/embriologia , Morfogênese/genética , Doenças Nasais/genética , beta Catenina/genética , beta Catenina/fisiologia , Animais , Adesão Celular , Coristoma/congênito , Olho/embriologia , Cristalino/citologia , Camundongos , Camundongos Transgênicos , Doenças Nasais/congênito , Transdução de Sinais/genética , Proteínas Wnt/fisiologia
10.
Int J Mol Med ; 18(1): 65-76, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16786157

RESUMO

Galectin-4 and its homologue galectin-6 are members of the tandem-repeat subfamily of monomer divalent galectins. Expression of mouse galectin-4 and galectin-6 by RT-PCR using primers designed to distinguish both galectin transcripts indicates that both are expressed in the small intestine, colon, liver, kidney, spleen and heart and P19X1 cells while only galectin-4 is expressed in BW-5147 and 3T3 cell lines. In situ hybridization confirmed the presence of galectin-4/-6 transcripts in the liver and small intestine. Galectin-4 is expressed in spermatozoons and oocytes and its expression during early mouse emryogenesis appears in 8-cell embryos and remains in later stages, as tested by RT-PCR. To study the role of carbohydrate recognition domains (CRDs) in oligosaccharide binding and epitope recognition, we cloned mouse full-length galectin-4 and galectin-6 cDNA and constructed bacterial expression vectors producing histidin-tagged recombinant galectin-4 and its truncated CRD1 and CRD2 forms. Oligosaccharide binding profile for all recombinant forms was assessed using Glycan Array available through the Consortium for Functional Glycomics. Acquired data indicate that mGalectin-4 binds to alpha-GalNAc and alpha-Gal A and B type structures with or without fucose. While the CRD2 domain has a high specificity and affinity for A type-2 alpha-GalNAc structures, the CRD1 domain has a broader specificity in correlation to the total binding profile. These data suggest that CRD2 might be the dominant binding domain of mouse galectin-4. Mapping of epitopes reactive for biotinylated his-tagged CRD1, CRD2 and mGalectin-4 performed on mouse cryosections showed that all three forms bind to alveolar macrophages, macrophages of red pulp of the spleen and proximal tubuli of the kidney and this binding was inhibited by 5 mM lactose. Interestingly, mGalectin-4, but not CRD forms, binds to the suprabasal layer of squamous epithelium of the tongue, suggesting that the link region also plays an important role in ligand recognition.


Assuntos
Epitopos/metabolismo , Galectina 4/genética , Galectinas/genética , Oligossacarídeos/metabolismo , Células 3T3 , Animais , Sítios de Ligação/genética , Sequência de Carboidratos , Linhagem Celular Tumoral , DNA Complementar/genética , Epitopos/química , Galectina 4/metabolismo , Galectinas/metabolismo , Perfilação da Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ/métodos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Oligossacarídeos/química , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...