Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 465(2): 119-129, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32697973

RESUMO

During ocular development, periocular neural crest cells (pNC) migrate into the region between the lens and presumptive corneal epithelium to form the corneal endothelium and stromal keratocytes. Although defects in neural crest cell development are associated with ocular dysgenesis, very little is known about the molecular mechanisms involved in this process. This study focuses on the corneal endothelium, a monolayer of specialized cells that are essential for maintaining normal hydration and transparency of the cornea. In avians, corneal endothelial cells are first to be specified from the pNC during their migration into the presumptive corneal region. To investigate the signals required for formation of the corneal endothelium, we utilized orthotopic and heterotopic injections of dissociated quail pNC into chick ocular regions. We find that pNC are multipotent and that the nascent cornea is competent to induce differentiation of ectopically injected pNC into corneal endothelium. Injected pNC downregulate expression of multipotency transcription factors and upregulate genes that are consistent with ontogenesis of the chick corneal endothelium. Importantly, we showed that TGFß2 is expressed by the nascent lens and the corneal endothelium, and that TGFß signaling plays a critical role in changing the molecular signature of pNC in vitro. Collectively, our results demonstrate the significance of the ocular environmental cues towards pNC differentiation, and have potential implications for clinical application of stem cells in the anterior segment.


Assuntos
Proteínas Aviárias/metabolismo , Endotélio Corneano/embriologia , Crista Neural/embriologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Embrião de Galinha , Galinhas , Endotélio Corneano/citologia , Crista Neural/citologia
2.
Invest Ophthalmol Vis Sci ; 57(3): 1072-81, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26968737

RESUMO

PURPOSE: The homeodomain transcription factor, PITX2, is at the apex of a genetic pathway required for corneal development, but the critical effector genes regulated by the PITX2 remain unknown. The purpose of this study was to discover and validate PITX2-dependent mechanisms required for specifying cell lineages and establishing angiogenic privilege within the developing cornea. METHODS: Microarrays were used to compare gene expression in corneas isolated from temporal Pitx2 knockout embryos and control littermates. Quantitative RT-PCR and immunohistochemistry was used to further validate Tfap2b expression differences in Pitx2 knockout versus control corneas. In situ hybridization and protein immunohistochemistry were used to assay eyes of a Tfap2b allelic series of embryos to identify differentiated cellular lineages in the cornea, blood vessel endothelium, or lymphatic vessel endothelium. RESULTS: We show that PITX2 is required for the expression of Tfap2b, encoding the AP-2ß transcription factor, in the neural crest during corneal development. Markers of differentiated corneal epithelium and stroma are expressed in the absence of AP-2ß. In contrast, markers of differentiated corneal endothelium are not expressed in the absence of AP-2ß. Endomucin+ blood vessels are present throughout the developing corneal stroma in the absence of AP-2ß, whereas LYVE1+ lymphatic vessels are not found. CONCLUSIONS: The AP-2ß transcription factor is an important effector of PITX2 function during corneal development, required for differentiation of corneal endothelium and establishment of angiogenic privilege. Unlike PITX2, AP-2ß is not required for the early expression of available lineage specific markers for the corneal epithelium and stroma during embryogenesis, nor establishment of lymphangiogenic privilege. Therefore, additional PITX2-dependent factors likely regulate these latter processes during embryonic development. These results extend our understanding of the genetic mechanisms regulating cornea development.


Assuntos
Endotélio Corneano/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Morfogênese/genética , Neovascularização Fisiológica/genética , Prenhez , Fator de Transcrição AP-2/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Córnea/embriologia , Córnea/metabolismo , Endotélio Corneano/metabolismo , Feminino , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Gravidez , Fator de Transcrição AP-2/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Proteína Homeobox PITX2
3.
Proc Natl Acad Sci U S A ; 108(7): 2819-24, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21285373

RESUMO

Primary cilia are required for several signaling pathways, but their function in cellular morphogenesis is poorly understood. Here we show that emergence of an hexagonal cellular pattern during development of the corneal endothelium (CE), a monolayer of neural crest-derived cells that maintains corneal transparency, depends on a precise temporal control of assembly of primary cilia that subsequently disassemble in adult corneal endothelial cells (CECs). However, cilia reassembly occurs rapidly in response to an in vivo mechanical injury and precedes basal body polarization and cellular elongation in mature CECs neighboring the wound. In contrast, CE from hypomorphic IFT88 mutants (Tg737(orpk)) or following in vivo lentiviral-mediated IFT88 knockdown display dysfunctional cilia and show disorganized patterning, mislocalization of junctional markers, and accumulation of cytoplasmic acetylated tubulin. Our results indicate an active role of cilia in orchestrating coordinated morphogenesis of CECs during development and repair and define the murine CE as a powerful in vivo system to study ciliary-based cellular dynamics.


Assuntos
Cílios/fisiologia , Perda de Células Endoteliais da Córnea/fisiopatologia , Endotélio Corneano/embriologia , Endotélio Corneano/lesões , Morfogênese , Animais , Endotélio Corneano/ultraestrutura , Técnicas de Silenciamento de Genes , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Interferência de RNA , Proteínas Supressoras de Tumor/genética
4.
Invest Ophthalmol Vis Sci ; 49(5): 1843-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18436818

RESUMO

PURPOSE: The zinc finger transcription factor Zeb1 binds to E-box-like sequences and is important for maintaining repression of epithelial specification genes in vivo. Overexpression of Zeb1 in cancer triggers epithelial-mesenchymal transition, which facilitates metastasis. The mutation of ZEB1 in humans is linked to posterior polymorphous corneal dystrophy (PPCD), in which an epithelial transition of the corneal endothelium is associated with abnormal endothelial proliferation. The purpose of this study is to determine whether Zeb1 null or heterozygous mice may provide an animal model for PPCD. METHODS: Corneal morphology, protein and mRNA expression, and cell proliferation were compared in wild-type and Zeb1 gene knockout mice by immunostaining, real-time PCR, and BrdU incorporation. mRNA expression in isolated embryo fibroblasts derived from wild-type, Zeb1 heterozygous, and null mice was analyzed by real-time PCR RESULTS: Zeb1 null mice late in gestation show ectopic expression of epithelial genes in the corneal endothelium and keratocytes, including the basement membrane component COL4A3, which is ectopically expressed by the corneal endothelium in PPCD. These embryos also show abnormal corneal endothelial and keratocyte proliferation, corneal thickening, and corneolenticular and iridocorneal adhesions. Adult Zeb1 heterozygous mice exhibit these same corneal defects. The ectopic expression of epithelial genes extended to embryonic fibroblasts derived from Zeb1 heterozygous and null mice, suggesting that Zeb1 may have a more general role in the suppression of an epithelial phenotype. CONCLUSIONS: The authors conclude that Zeb1 heterozygous and null mice show features of PPCD and thus should provide an animal model for genetic dissection of pathways contributing to the disease.


Assuntos
Distrofias Hereditárias da Córnea/genética , Modelos Animais de Doenças , Endotélio Corneano/metabolismo , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Mutação/fisiologia , Dedos de Zinco/fisiologia , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Bromodesoxiuridina/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proliferação de Células , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Distrofias Hereditárias da Córnea/embriologia , Distrofias Hereditárias da Córnea/patologia , Endotélio Corneano/embriologia , Endotélio Corneano/patologia , Epitélio Corneano/embriologia , Epitélio Corneano/metabolismo , Epitélio Corneano/patologia , Feminino , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Dosagem de Genes , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homeobox 1 de Ligação a E-box em Dedo de Zinco
5.
Development ; 127(3): 533-42, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10631174

RESUMO

The anterior segment of the vertebrate eye is constructed by proper spatial development of cells derived from the surface ectoderm, which become corneal epithelium and lens, neuroectoderm (posterior iris and ciliary body) and cranial neural crest (corneal stroma, corneal endothelium and anterior iris). Although coordinated interactions between these different cell types are presumed to be essential for proper spatial positioning and differentiation, the requisite intercellular signals remain undefined. We have generated transgenic mice that express either transforming growth factor (alpha) (TGF(alpha)) or epidermal growth factor (EGF) in the ocular lens using the mouse (alpha)A-crystallin promoter. Expression of either growth factor alters the normal developmental fate of the innermost corneal mesenchymal cells so that these cells often fail to differentiate into corneal endothelial cells. Both sets of transgenic mice subsequently manifest multiple anterior segment defects, including attachment of the iris and lens to the cornea, a reduction in the thickness of the corneal epithelium, corneal opacity, and modest disorganization in the corneal stroma. Our data suggest that formation of a corneal endothelium during early ocular morphogenesis is required to prevent attachment of the lens and iris to the corneal stroma, therefore permitting the normal formation of the anterior segment.


Assuntos
Câmara Anterior/anormalidades , Câmara Anterior/embriologia , Ectoderma/fisiologia , Endotélio Corneano/embriologia , Fator de Crescimento Epidérmico/fisiologia , Cristalino/embriologia , Fator de Crescimento Transformador alfa/fisiologia , Animais , Caderinas/análise , Caderinas/genética , Cristalinas/genética , Modelos Animais de Doenças , Desenvolvimento Embrionário e Fetal , Endotélio Corneano/anormalidades , Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Fator de Crescimento Transformador alfa/genética
6.
J Cataract Refract Surg ; 20(6): 638-42, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7837076

RESUMO

Using an organotypic culture method, we evaluated the biocompatibility of two kinds of intraocular lenses: conventional poly(methyl methacrylate) (PMMA) and heparin-surface-modified (HSM) PMMA. Chicken corneal endothelium from embryos incubated for 14 days was placed on an agar medium and covered with one of three types of materials: PMMA, HSM PMMA, and a control, Thermanox. Experiments at five different times measured cell migration, cell multiplication, and cell adhesion. Scanning electron microscopy showed a low level of corneal endothelial adhesion on the HSM IOL surface. There was a significant difference between the HSM and untreated lenses in migration surface and cell density, with the HSM lenses having better biocompatibility.


Assuntos
Materiais Biocompatíveis , Endotélio Corneano/ultraestrutura , Heparina , Lentes Intraoculares , Animais , Adesão Celular , Divisão Celular , Movimento Celular , Embrião de Galinha , Endotélio Corneano/embriologia , Endotélio Corneano/fisiologia , Teste de Materiais , Metilmetacrilato , Metilmetacrilatos , Técnicas de Cultura de Órgãos/métodos
7.
Cornea ; 9(4): 312-7, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1706651

RESUMO

Flat mounts of human corneal endothelial cells (HCECs) were examined immunohistochemically by using a wide assortment of monoclonal antibodies against the five classes of intermediate filaments (IFs) and actin and myosin. HCECs showed uniform immunostaining with monoclonal antibodies against the 40-kD (CK 19) and 45-kD (CK 18) cytokeratin (CK). Only part of the endothelial cells reacted with monoclonal antibodies against the 52-kD (CK 8) and 54-kD (CK 7) cytokeratin polypeptides and with monoclonal antibodies against vimentin. Monoclonal antibodies against the low- and middle-molecular-mass neurofilament proteins produced positive staining of all HCECs. No positivity was obtained with antibodies against desmin or glial fibrillary acidic protein. In addition, positive immunostaining with monoclonal antibodies against actin and slow myosin demonstrate that these proteins form part of the cytoskeleton of HCECs. The results of this study show that immunostaining of flat cell preparations is very useful for studies on HCECs. HCECs display an unusual combination of cytokeratin IFs and neurofilaments, together with vimentin, and are heterogeneous with respect to their IF makeup. These findings are discussed in relation to the presumed origin of HCECs.


Assuntos
Citoesqueleto , Endotélio Corneano/ultraestrutura , Actinas/análise , Anticorpos Monoclonais , Desmina/análise , Endotélio Corneano/química , Endotélio Corneano/embriologia , Humanos , Técnicas Imunoenzimáticas , Filamentos Intermediários/química , Queratinas/análise , Miosinas/análise , Vimentina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA