Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Exp Eye Res ; 200: 108201, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32888962

RESUMEN

Mesenchymal stromal cells (MSC), with progenitor cell and immunological properties, have been cultivated from numerous vascularized tissues including bone marrow, adipose tissue and the corneal-limbus of the eye. After observing mesenchymal cells as contaminants in primary cultures of vascular endothelial cells derived from the choroidal tunic of the human eye, we investigated whether the choroid might also provide a source of cultured MSC. Moreover, we examined the effect of the choroidal stromal cells (Ch-SC) on the proliferation of freshly isolated choroidal vascular endothelial cells (ChVEC) in vitro. The phenotype of cultures established from five choroidal tissue donors was examined by flow cytometry and immunocytochemistry. The potential for mesenchymal cell differentiation was examined in parallel with MSC established from human bone marrow. Additional cultures were growth-arrested by treatment with mitomycin-C, before being tested as a potential feeder layer for ChVEC. The five unique cultures established from choroidal stroma displayed a phenotype consistent with the accepted definition for MSC (CD34-, CD45-, HLA-DR-, CD73+, CD90+, and CD105+), including the capacity for mesenchymal differentiation when cultivated under osteogenic, adipogenic and chondrogenic conditions. Growth-arrested Ch-SC inhibited the proliferation of ChVEC derived from five separate donors. Cultures of Ch-SC secreted approximately 40-fold higher concentrations of the anti-angiogenic factor pigment epithelium derived factor (PEDF/serpin F1) compared to the pro-angiogenic factor, vascular endothelial growth factor (VEGF), regardless of normal or growth-arrested state. Our results provide first evidence of a resident MSC cell type within the choroid and encourage investigation of new mechanisms for altering the growth of ChVEC.


Asunto(s)
Coroides/irrigación sanguínea , Células Endoteliales/citología , Endotelio Vascular/citología , Células Madre Mesenquimatosas/citología , Células del Estroma/citología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Coroides/citología , Citometría de Flujo , Humanos , Fenotipo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Exp Eye Res ; 173: 24-31, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29680447

RESUMEN

Research is currently under way to produce tissue engineered corneal endothelium transplants for therapeutic use in humans. This work requires the use of model animals, both for the supply of corneal endothelial cells (CECs) for experimentation, and to serve as recipients for test transplants. A variety of species can be used, however, a number of important advantages can be gained by using sheep as transplant recipients. The purpose of the present study was therefore to develop a method for culturing sheep CECs that would be suitable for the eventual construction of corneal endothelium grafts destined for sheep subjects. A method was established for culturing sheep CECs and these were compared to cultured human CECs. Results showed that cultured sheep and human CECs had similar growth characteristics when expanded from corneal endothelium explants on gelatin-coated plates, and achieved similar cell densities after several weeks. Furthermore, the markers zonula occludens-1, N-cadherin and sodium potassium ATPase could be immunodetected in similar staining patterns at cell boundaries of cultured CECs from both species. This work represents the first detailed study of sheep CEC cultures, and is the first demonstration of their similarities to human CEC cultures. Our results indicate that sheep CECs would be an appropriate substitute for human CECs when developing methods to produce tissue engineered corneal endothelium transplants.


Asunto(s)
Técnicas de Cultivo de Célula , Endotelio Corneal/citología , Adolescente , Animales , Biomarcadores/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Recuento de Células , Células Cultivadas , Endotelio Corneal/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Ovinos , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ingeniería de Tejidos , Proteína de la Zonula Occludens-1/metabolismo
3.
Exp Eye Res ; 170: 92-100, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29476773

RESUMEN

The corneal endothelium is a monolayer of epithelial cells that lines the posterior surface of the cornea and is essential for maintenance of corneal transparency. Wound healing within the corneal endothelium typically occurs through cell spreading and migration rather than through proliferation. The mechanisms that control corneal endothelial cell migration are unclear. In this study we demonstrate that cultures of corneal endothelial cells display reduced migration in scratch wound assays, and reduced levels of E-cadherin mRNA, following suppression of ligand-activated Eph receptor signalling by treatment with lithocholic acid. Two Eph receptors, EphA1 and EphA2, were subsequently detected in corneal endothelial cells, and their potential involvement during migration was explored through gene silencing using siRNAs. EphA2 siRNA reduced levels of mRNA for both EphA2 and N-cadherin, but increased levels of mRNA for both EphA1 and E-cadherin. No effect, however, was observed for EphA2 siRNA on migration. Our results indicate a potential role for Eph receptor signalling during corneal endothelial cell migration via changes in cadherin expression. Nevertheless, defining a precise role for select Eph receptors is likely to be complicated by crosstalk between Eph-mediated signalling pathways.


Asunto(s)
Movimiento Celular/fisiología , Endotelio Corneal/citología , Receptor EphA1/fisiología , Receptor EphA2/fisiología , Transducción de Señal/fisiología , Animales , Cadherinas/metabolismo , Línea Celular , Supervivencia Celular , Detergentes/farmacología , Endotelio Corneal/efectos de los fármacos , Endotelio Corneal/metabolismo , Efrina-A1/genética , Efrina-A1/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Silenciador del Gen , Humanos , Ácido Litocólico/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor EphA1/antagonistas & inhibidores , Receptor EphA2/antagonistas & inhibidores , Ovinos
4.
Exp Eye Res ; 176: 196-206, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30025918

RESUMEN

In response to an unexpected observation of apparent localisation by immunocytochemistry, we have investigated the potential expression and function of P-selectin (CD62P) in human corneal epithelial cells. The SV40 immortalised cell line, HCE-T (validated by STR profiling), along with multiple donor corneal-limbal tissue samples, were examined for P-selectin expression using a combination of immunocytochemistry, Western blotting, RT-PCR and immunohistochemistry. Potential expression of the major ligand for P-selectin (P-selectin glycoprotein ligand-1; PSGL-1; CD162) was also examined by immunocytochemistry and RT-PCR. A selective inhibitor of P-selectin-PSGL-1 binding (KF38789) was subsequently tested for effects on HCE-T cells using a cell culture gap-closure assay. HCE-T cells as well as primary epithelial cultures derived from donor corneal-limbal tissue, displayed positive immunostaining for P-selectin. Staining was particularly evident at cell-cell boundaries and at the outer edge of expanding epithelial islands. P-selectin expression was confirmed by Western blotting and RT-PCR (validated by product sequencing), as well as by immunohistochemistry performed on serial sections of corneal-limbal tissue stained for P-selectin, keratin 3 and p63. PSGL-1 was detected by RT-PCR and immunocytochemistry in both corneal epithelial cells as well as human limbal fibroblasts (HLF). KF38789 (5 µM) significantly reduced closure of a 500-µm gap between confluent sheets of HCE-T cells over an 8-hr period (by ∼40%, p < 0.01; paired two-tailed T test), but had no effect on culture gap-closure by either HLF or murine 3T3 fibroblasts. These results provide evidence of P-selectin expression in human corneal epithelial cells and suggest a potential role for this glycoprotein in facilitating the net movement of confluent sheets of human corneal epithelial cells.


Asunto(s)
Epitelio Corneal/metabolismo , Selectina-P/genética , Selectina-P/metabolismo , Biomarcadores/metabolismo , Western Blotting , Separación Celular/métodos , Células Cultivadas , Fibroblastos/metabolismo , Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Limbo de la Córnea/citología , Glicoproteínas de Membrana/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Exp Eye Res ; 147: 1-11, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27085211

RESUMEN

The New Zealand White rabbit has been widely used as a model of limbal stem cell deficiency (LSCD). Current techniques for experimental induction of LSCD utilize caustic chemicals, or organic solvents applied in conjunction with a surgical limbectomy. While generally successful in depleting epithelial progenitors, the depth and severity of injury is difficult to control using chemical-based methods. Moreover, the anterior chamber can be easily perforated while surgically excising the corneal limbus. In the interest of creating a safer and more defined LSCD model, we have therefore evaluated a mechanical debridement technique based upon use of the AlgerBrush II rotating burr. An initial comparison of debridement techniques was conducted in situ using 24 eyes in freshly acquired New Zealand White rabbit cadavers. Techniques for comparison (4 eyes each) included: (1) non-wounded control, (2) surgical limbectomy followed by treatment with 100% (v/v) n-heptanol to remove the corneal epithelium (1-2 min), (3) treatment of both limbus and cornea with n-heptanol alone, (4) treatment of both limbus and cornea with 20% (v/v) ethanol (2-3 min), (5) a 2.5-mm rounded burr applied to both the limbus and cornea, and (6) a 1-mm pointed burr applied to the limbus, followed by the 2.5-mm rounded burr applied to the cornea. All corneas were excised and processed for histology immediately following debridement. A panel of four assessors subsequently scored the degree of epithelial debridement within the cornea and limbus using masked slides. The 2.5-mm burr most consistently removed the corneal and limbal epithelia. Islands of limbal epithelial cells were occasionally retained following surgical limbectomy/heptanol treatment, or use of the 1-mm burr. Limbal epithelial cells were consistently retained following treatment with either ethanol or n-heptanol alone, with ethanol being the least effective treatment overall. The 2.5-mm burr method was subsequently evaluated in the right eye of 3 live rabbits by weekly clinical assessments (photography and slit lamp examination) for up to 5 weeks, followed by histological analyses (hematoxylin & eosin stain, periodic acid-Schiff stain and immunohistochemistry for keratin 3 and 13). All 3 eyes that had been completely debrided using the 2.5-mm burr displayed symptoms of ocular surface failure as defined by retention of a prominent epithelial defect (∼40% of corneal surface at 5 weeks), corneal neovascularization (2-3 quadrants), reduced corneal transparency and conjunctivalization of the corneal surface (demonstrated by the presence of goblet cells and/or staining for keratin 13). In conclusion, our findings indicate that the AlgerBrush II rotating burr is an effective method for the establishment of ocular surface failure in New Zealand White rabbits. In particular, we recommend use of the 2.5-mm rotating burr for improved efficiency of epithelial debridement and safety compared to surgical limbectomy.


Asunto(s)
Córnea/cirugía , Desbridamiento/instrumentación , Modelos Animales de Enfermedad , Epitelio Corneal/cirugía , Equipo Quirúrgico , Animales , Córnea/patología , Desbridamiento/métodos , Femenino , Limbo de la Córnea/patología , Limbo de la Córnea/cirugía , Conejos , Células Madre/citología
6.
Exp Eye Res ; 127: 9-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25035050

RESUMEN

The routine cultivation of human corneal endothelial cells, with the view to treating patients with endothelial dysfunction, remains a challenging task. While progress in this field has been buoyed by the proposed existence of progenitor cells for the corneal endothelium at the corneal limbus, strategies for exploiting this concept remain unclear. In the course of evaluating methods for growing corneal endothelial cells, we have noted a case where remarkable growth was achieved using a serial explant culture technique. Over the course of 7 months, a single explant of corneal endothelium, acquired from cadaveric human tissue, was sequentially seeded into 7 culture plates and on each occasion produced a confluent cell monolayer. Sample cultures were confirmed as endothelial in origin by positive staining for glypican-4. On each occasion, small cells, closest to the tissue explant, developed into a highly compact layer with an almost homogenous structure. This layer was resistant to removal with trypsin and produced continuous cell outgrowth during multiple culture periods. The small cells gave rise to larger cells with phase-bright cell boundaries and prominent immunostaining for both nestin and telomerase. Nestin and telomerase were also strongly expressed in small cells immediately adjacent to the wound site, following transfer of the explant to another culture plate. These findings are consistent with the theory that progenitor cells for the corneal endothelium reside within the limbus and provide new insights into expected expression patterns for nestin and telomerase within the differentiation pathway.


Asunto(s)
Endotelio Corneal/citología , Limbo de la Córnea/citología , Células Madre/citología , Adulto , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Separación Celular/métodos , Endotelio Corneal/metabolismo , Glipicanos/metabolismo , Humanos , Nestina/metabolismo , Fenotipo , Células Madre/metabolismo
7.
J Vis Exp ; (156)2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32090992

RESUMEN

Corneal endothelial cell cultures have a tendency to undergo epithelial-to-mesenchymal transition (EMT) after loss of cell-to-cell contact. EMT is deleterious for the cells as it reduces their ability to form a mature and functional layer. Here, we present a method for establishing and subculturing human and sheep corneal endothelial cell cultures that minimizes the loss of cell-to-cell contact. Explants of corneal endothelium/Descemet's membrane are taken from donor corneas and placed into tissue culture under conditions that allow the cells to collectively migrate onto the culture surface. Once a culture has been established, the explants are transferred to fresh plates to initiate new cultures. Dispase II is used to gently lift clumps of cells off tissue culture plates for subculturing. Corneal endothelial cell cultures that have been established using this protocol are suitable for transferring to biomaterial membranes to produce tissue-engineered cell layers for transplantation in animal trials. A custom-made device for supporting biomaterial membranes during tissue culture is described and an example of a tissue-engineered graft composed of a layer of corneal endothelial cells and a layer of corneal stromal cells on either side of a collagen type I membrane is presented.


Asunto(s)
Materiales Biocompatibles/farmacología , Lámina Limitante Posterior/metabolismo , Células Endoteliales/citología , Endotelio Corneal/crecimiento & desarrollo , Animales , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Lámina Limitante Posterior/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Ovinos , Donantes de Tejidos , Proteína de la Zonula Occludens-1/metabolismo
8.
Cancer Res ; 67(10): 4751-8, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510403

RESUMEN

Cutaneous squamous cell carcinomas (CSCC) are a common malignancy of keratinocytes that arise in sites of the skin exposed to excessive UV radiation. In the present study, we show that human SCC cell lines, preneoplastic solar keratoses (SK), and CSCC are associated with perturbations in glutathione peroxidase (GPX) activity and peroxide levels. Specifically, we found that two of three SKs and four of five CSCCs, in vivo, were associated with decreased GPX activity and all SKs and CSCCs were associated with an elevated peroxide burden. Given the association of decreased GPX activity with CSCC, we examined the basis for the GPX deficiency in the CSCCs. Our data indicated that GPX was inactivated by a post-translational mechanism and that GPX could be inactivated by increases in intracellular peroxide levels. We next tested whether the decreased peroxidase activity coupled with an elevated peroxidative burden might contribute to CSCC formation in vivo. This was tested in Gpx1(-/-) and Gpx2(-/-) mice exposed to solar-simulated UV radiation. These studies showed that Gpx2 deficiency predisposed mice to UV-induced CSCC formation. These results suggest that inactivation of GPX2 in human skin may be an early event in UV-induced SCC formation.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/etiología , Glutatión Peroxidasa/metabolismo , Neoplasias Inducidas por Radiación/enzimología , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/etiología , Activación Enzimática , Glutatión Peroxidasa/biosíntesis , Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/genética , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Queratinocitos/enzimología , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Neoplasias Inducidas por Radiación/etiología , Peróxidos/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Rayos Ultravioleta
9.
Cell Transplant ; 28(9-10): 1257-1270, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31208228

RESUMEN

While limbal epithelial cells are used for treating ocular surface wounds, the therapeutic potential of mesenchymal cells cultivated from the limbal stroma (LMSC) is less clear. We have therefore examined the effects of LMSC when applied to acute ocular surface wounds. LMSC derived from male rabbits (RLMSC) were applied to the ocular surface of female rabbits immediately following removal of the corneal and limbal epithelium. Human amniotic membrane (HAM) was used as the vehicle for implanting the RLMSC. The effects of RLMSC were examined when applied alone (n = 3) and in conjunction with a stratified culture of human limbal epithelial cells (HLE) grown on the opposing surface of the HAM (n = 3). Outcomes were monitored over 3 months in comparison with animals receiving no treatment (n = 3) or treatment with HLE alone on HAM (n = 3). Animals treated with RLMSC (n = 6) displayed faster re-epithelialization (∼90% versus 70% healing after 12 weeks), with best results being observed when RLMSC were pre-cultivated and implanted in the presence of HLE (p < 0.01; 90% healing by 7 weeks). While all animals displayed conjunctival cells on the corneal surface (by presence of goblet cells and/or keratin 13 expression) and corneal neovascularization, evidence of corneal epithelial regeneration was observed in animals that received RLMSC in the presence of HLE (by staining for keratin 3 and the absence of goblet cells). Conversely, corneal neovascularization was significantly greater when RLMSC were applied in the absence of HLE (<0.05; 90% of cornea compared with 20-30% in other cohorts). Nevertheless, neither human nuclear antigen nor rabbit Y chromosome were detected within the regenerated epithelium. Our results demonstrate that while cultured LMSC encourage corneal re-epithelialization, healing is improved by the pre-cultivation and implantation of these mesenchymal cells in the presence of limbal epithelial cells.


Asunto(s)
Células Epiteliales , Epitelio Corneal , Lesiones Oculares , Limbo de la Córnea , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Cicatrización de Heridas , Enfermedad Aguda , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio Corneal/lesiones , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Lesiones Oculares/metabolismo , Lesiones Oculares/patología , Lesiones Oculares/terapia , Femenino , Humanos , Limbo de la Córnea/lesiones , Limbo de la Córnea/metabolismo , Limbo de la Córnea/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Conejos
10.
Mater Sci Eng C Mater Biol Appl ; 105: 110131, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31546376

RESUMEN

Silk fibroin membrane displays potential for ocular tissue reconstruction as demonstrated by its ability to support a functioning retinal pigment epithelium (RPE) in vitro. Nevertheless, translation of these findings to the clinic will require the use of membranes that can be readily handled and implanted into diseased retinas, with minimal impact on the surrounding healthy tissue. To this end, we optimized the physical properties of fibroin membranes to enable surgical handling during implantation into the retina, without compromising biocompatibility or permeability. Our central hypothesis is that optimal strength and permeability can be achieved by combining the porogenic properties of poly(ethylene glycol) (PEG) with the crosslinking properties of horseradish peroxidase (HRP). Our study reveals that PEG used in conjunction with HRP enables the production of fibroin membranes with superior handling properties to conventional fibroin membranes. More specifically, the modified membranes could be more easily implanted into the retinas of rats and displayed good evidence of biocompatibility. Moreover, the modified membranes retained the ability to support construction of functional RPE derived from pluripotent stem cells. These findings pave the way for preclinical studies of RPE-implantation using the optimized fibroin membranes.


Asunto(s)
Fibroínas/química , Membranas Artificiales , Prótesis Visuales , Animales , Bombyx , Células Madre Embrionarias Humanas/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Permeabilidad , Fagocitosis , Ratas , Epitelio Pigmentado de la Retina/citología , Soluciones , Espectroscopía Infrarroja por Transformada de Fourier , Resistencia a la Tracción
11.
Curr Biol ; 12(13): 1117-23, 2002 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-12121619

RESUMEN

The hindbrain (brainstem) of all vertebrates follows a segmental developmental strategy and has been the focus of intense study not only for its intrinsic interest but also as a model for how more complex regions of the brain are patterned. Segmentation ultimately serves to organize the development of neuronal populations and their projections, and regional diversity is achieved through each segment having its own identity. The latter being established through differential expression of a hierarchy of transcription factors, including Hox genes, Krox20, and Kreisler/Valentino. Here we identify a novel signaling center in the zebrafish embryo that arises prior to establishment of segmental patterning and which is located centrally within the hindbrain territory in a region that corresponds to the presumptive rhombomere 4. We show that signaling from this region by two members of the FGF family of secreted proteins, FGF3 and FGF8, is required to establish correct segmental identity throughout the hindbrain and for subsequent neuronal development. Spatiotemporal studies of Fgf expression suggest that this patterning mechanism is conserved during hindbrain development in other vertebrate classes.


Asunto(s)
Tipificación del Cuerpo/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Rombencéfalo/embriología , Transducción de Señal , Proteínas de Pez Cebra , Animales , Proteínas de Unión al ADN/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz , Efrina-B2/biosíntesis , Factor 3 de Crecimiento de Fibroblastos , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología
12.
Cell Transplant ; 26(11): 1717-1732, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29338382

RESUMEN

When used as scaffolds for cell therapies, biomaterials often present basic handling and logistical problems for scientists and surgeons alike. The quest for an appropriate mounting device for biomaterials is therefore a significant and common problem. In this review, we provide a detailed overview of the factors to consider when choosing an appropriate mounting device including those experienced during cell culture, quality assurance, and surgery. By way of example, we draw upon our combined experience in developing epithelial cell therapies for the treatment of eye diseases. We discuss commercially available options for achieving required goals and provide a detailed analysis of 4 experimental designs developed within our respective laboratories in Australia, the United Kingdom, and Belgium.


Asunto(s)
Materiales Biocompatibles/química , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Amnios/citología , Humanos , Oftalmología
13.
Stem Cells Int ; 2016: 8310127, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27648078

RESUMEN

Scaffolds prepared from silk fibroin derived from cocoons of the domesticated silkworm moth Bombyx mori have demonstrated potential to support the attachment and growth of human limbal epithelial (HLE) cells in vitro. In this study, we attempted to further optimize protocols to promote the expansion of HLE cells on B. mori silk fibroin- (BMSF-) based scaffolds. BMSF films were initially coated with different extracellular matrix proteins and then analysed for their impact on corneal epithelial cell adhesion, cell morphology, and culture confluency. Results showed that collagen I, collagen III, and collagen IV consistently improved HCE-T cell adherence, promoted an elongated cell morphology, and increased culture confluency. By contrast, ECM coating had no significant effect on the performance of primary HLE cells cultured on BMSF films. In the second part of this study, primary HLE cells were grown on BMSF films in the presence of medium (SHEM) supplemented with keratinocyte growth factor (KGF) and the Rho kinase inhibitor, Y-27632. The results demonstrated that SHEM medium supplemented with KGF and Y-27632 dramatically increased expression of corneal differentiation markers, keratin 3 and keratin 12, whereas expression of the progenitor marker, p63, did not appear to be significantly influenced by the choice of culture medium.

14.
Dev Dyn ; 237(3): 592-601, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18224711

RESUMEN

In the head, neural crest cells generate ectomesenchymal derivatives: cartilage, bone, and connective tissue. Indeed, these cells generate much of the cranial skeleton. There have, however, been few studies of how this lineage is established. Here, we show that neural crest cells stop expressing early neural crest markers upon entering the pharyngeal arches and switch to become ectomesenchymal. By contrast, those neural crest cells that do not enter the arches persist in their expression of early neural crest markers. We further show that fibroblast growth factor (FGF) signaling is involved in directing neural crest cells to become ectomesenchymal. If neural crest cells are rendered insensitive to FGFs, they persist in their expression of early neural crest markers, even after entering the pharyngeal arches. However, our results further suggest that, although FGF signaling is required for the realization of the ectomesenchymal lineages, other cues from the pharyngeal epithelia are also likely to be involved.


Asunto(s)
Región Branquial/embriología , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/metabolismo , Mesodermo/fisiología , Cresta Neural/embriología , Animales , Antígenos de Superficie/metabolismo , Región Branquial/citología , Embrión de Pollo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/citología , Transducción de Señal , Pez Cebra/embriología
15.
Dev Biol ; 264(2): 522-36, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14651935

RESUMEN

Characterisation of human craniofacial syndromes and studies in transgenic mice have demonstrated the requirement for Fgf signalling during morphogenesis of membrane bone of the cranium. Here, we report that Fgf activity is also required for development of the oro-pharyngeal skeleton, which develops first as cartilage with some elements subsequently becoming ossified. We show that inhibition of FGF receptor activity in the zebrafish embryo following neural crest emigration from the neural tube results in complete absence of neurocranial and pharyngeal cartilages. Moreover, this Fgf signal is required during a 6-h period soon after initiation of neural crest migration. The spatial and temporal expression of Fgf3 and Fgf8 in pharyngeal endoderm and ventral forebrain and its correlation with patterns of Fgf signalling activity in migrating neural crest makes them candidate regulators of cartilage development. Inhibition of Fgf3 results in the complete absence of cartilage elements that normally form in the third, fourth, fifth, and sixth pharyngeal arches, while those of the first, second, and seventh arches are largely unaffected. Inhibition of Fgf8 alone has variable, but mild, effects. However, inhibition of both Fgf3 and Fgf8 together causes a complete absence of pharyngeal cartilages and the near-complete loss of the neurocranial cartilage. These data implicate Fgf3 and Fgf8 as key regulators of cartilage formation in the vertebrate head.


Asunto(s)
Cartílago/embriología , Factores de Crecimiento de Fibroblastos/fisiología , Cabeza/embriología , Faringe/embriología , Proteínas Proto-Oncogénicas/fisiología , Proteínas de Pez Cebra , Pez Cebra/embriología , Animales , Femenino , Factor 3 de Crecimiento de Fibroblastos , Factor 8 de Crecimiento de Fibroblastos , Transducción de Señal
16.
Development ; 130(18): 4337-49, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12900450

RESUMEN

Complex spatiotemporal expression patterns of fgf3 and fgf8 within the developing zebrafish forebrain suggest their involvement in its regionalisation and early development. These factors have unique and combinatorial roles during development of more posterior brain regions, and here we report similar findings for the developing forebrain. We show that Fgf8 and Fgf3 regulate different aspects of telencephalic development, and that Fgf3 alone is required for the expression of several telencephalic markers. Within the diencephalon, Fgf3 and Fgf8 act synergistically to pattern the ventral thalamus, and are implicated in the regulation of optic stalk formation, whereas loss of Fgf3 alone results in defects in ZLI development. Forebrain commissure formation was abnormal in the absence of either Fgf3 or Fgf8; however, most severe defects were observed in the absence of both. Defects were observed in patterning of both the midline territory, within which the commissures normally form, and neuronal populations, whose axons comprise the commissures. Analysis of embryos treated with an FGFR inhibitor suggests that continuous FGF signalling is required from gastrulation stages for normal forebrain patterning, and identifies additional requirements for FGFR activity.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Prosencéfalo/embriología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Pez Cebra , Pez Cebra/embriología , Animales , Axones/fisiología , Biomarcadores , Tipificación del Cuerpo , Muerte Celular , Diferenciación Celular , División Celular , Factor 3 de Crecimiento de Fibroblastos , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Hibridación in Situ , Morfogénesis , Oligonucleótidos Antisentido/metabolismo , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/fisiología , Proteínas Proto-Oncogénicas/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología
17.
Development ; 129(9): 2099-108, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11959820

RESUMEN

Fgf3 has long been implicated in otic placode induction and early development of the otocyst; however, the results of experiments in mouse and chick embryos to determine its function have proved to be conflicting. In this study, we determined fgf3 expression in relation to otic development in the zebrafish and used antisense morpholino oligonucleotides to inhibit Fgf3 translation. Successful knockdown of Fgf3 protein was demonstrated and this resulted in a reduction of otocyst size together with reduction in expression of early markers of the otic placode. fgf3 is co-expressed with fgf8 in the hindbrain prior to otic induction and, strikingly, when Fgf3 morpholinos were co-injected together with Fgf8 morpholinos, a significant number of embryos failed to form otocysts. These effects were made manifest at early stages of otic development by an absence of early placode markers (pax2.1 and dlx3) but were not accompanied by effects on cell division or death. The temporal requirement for Fgf signalling was established as being between 60% epiboly and tailbud stages using the Fgf receptor inhibitor SU5402. However, the earliest molecular event in induction of the otic territory, pax8 expression, did not require Fgf signalling, indicating an inductive event upstream of signalling by Fgf3 and Fgf8. We propose that Fgf3 and Fgf8 are required together for formation of the otic placode and act during the earliest stages of its induction.


Asunto(s)
Oído Interno/embriología , Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas de Pez Cebra , Pez Cebra/embriología , Pez Cebra/fisiología , Animales , Apoptosis , Secuencia de Bases , División Celular , Embrión de Pollo , Oído Interno/anomalías , Inducción Embrionaria/efectos de los fármacos , Inducción Embrionaria/genética , Inducción Embrionaria/fisiología , Factor 3 de Crecimiento de Fibroblastos , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Oligodesoxirribonucleótidos Antisentido/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA