Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Eye Res ; 187: 107772, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31445001

RESUMO

Mechanisms controlling the spatial configuration of the remarkably ordered collagen-rich extracellular matrix of the transparent cornea remain incompletely understood. We previously described the assembly of the emerging corneal matrix in the mid and late stages of embryogenesis and concluded that collagen fibril organisation was driven by cell-directed mechanisms. Here, the early stages of corneal morphogenesis were examined by serial block face scanning electron microscopy of embryonic chick corneas starting at embryonic day three (E3), followed by a Fourier transform analysis of three-dimensional datasets and theoretical considerations of factors that influence matrix formation. Eyes developing normally and eyes that had the lens surgically removed at E3 were studied. Uniformly thin collagen fibrils are deposited by surface ectoderm-derived corneal epithelium in the primary stroma of the developing chick cornea and form an acellular matrix with a striking micro-lamellar orthogonal arrangement. Fourier transform analysis supported this observation and indicated that adjacent micro-lamellae display a clockwise rotation of fibril orientation, depth-wise below the epithelium. We present a model which attempts to explain how, in the absence of cells in the primary stroma, collagen organisation might be influenced by cell-independent, intrinsic mechanisms, such as fibril axial charge derived from associated proteoglycans. On a supra-lamellar scale, fine cords of non-collagenous filamentous matrix were detected over large tissue volumes. These extend into the developing cornea from the epithelial basal lamina and appear to associate with the neural crest cells that migrate inwardly to form, first the corneal endothelium and then keratocytes which synthesise the mature, secondary corneal stroma. In a small number of experimental specimens, matrix cords were present even when periocular neural crest cell migration and corneal morphogenesis had been perturbed following removal of the lens at E3.


Assuntos
Córnea/embriologia , Matriz Extracelular/ultraestrutura , Animais , Embrião de Galinha , Sulfatos de Condroitina/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Córnea/metabolismo , Córnea/ultraestrutura , Substância Própria/embriologia , Substância Própria/metabolismo , Substância Própria/ultraestrutura , Dermatan Sulfato/metabolismo , Matriz Extracelular/metabolismo , Análise de Fourier , Imageamento Tridimensional , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Morfogênese/fisiologia
2.
Dev Biol ; 423(1): 77-91, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28095300

RESUMO

The C-X-C motif ligand 14 (CXCL14) is a recently discovered chemokine that is highly conserved in vertebrates and expressed in various embryonic and adult tissues. CXCL14 signaling has been implicated to function as an antiangiogenic and anticancer agent in adults. However, its function during development is unknown. We previously identified novel expression of CXCL14 mRNA in various ocular tissues during development. Here, we show that CXCL14 protein is expressed in the anterior eye at a critical time during neurovascular development and in the retina during neurogenesis. We report that RCAS-mediated knockdown of CXCL14 causes severe neural defects in the eye including precocious and excessive innervation of the cornea and iris. Absence of CXCL14 results in the malformation of the neural retina and misprojection of the retinal ganglion neurons. The ocular neural defects may be due to loss of CXCL12 modulation since recombinant CXCL14 diminishes CXCL12-induced axon growth in vitro. Furthermore, we show that knockdown of CXCL14 causes neovascularization of the cornea. Altogether, our results show for the first time that CXCL14 plays a critical role in modulating neurogenesis and inhibiting ectopic vascularization of the cornea during ocular development.


Assuntos
Padronização Corporal , Quimiocinas CXC/metabolismo , Olho/embriologia , Olho/metabolismo , Técnicas de Silenciamento de Genes , Sistema Nervoso/irrigação sanguínea , Sistema Nervoso/embriologia , Animais , Padronização Corporal/genética , Galinhas , Córnea/inervação , Córnea/metabolismo , Substância Própria/metabolismo , Epitélio Corneano/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Iris/embriologia , Iris/inervação , Modelos Biológicos , Codorniz , RNA Interferente Pequeno/metabolismo , Retina/patologia , Nervo Trigêmeo/embriologia , Nervo Trigêmeo/metabolismo
3.
Dev Biol ; 411(1): 128-39, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26783882

RESUMO

The anterior eye is comprised of an avascular cornea surrounded by a dense periocular vascular network and therefore serves as an excellent model for angiogenesis. Although signaling through PlexinD1 underlies various vascular patterning events during embryonic development, its role during the formation of the periocular vascular network is yet to be determined. Our recent study showed that PlexinD1 mRNA is expressed by periocular angioblasts and blood vessels during ocular vasculogenesis in patterns that suggest its involvement with Sema3 ligands that are concurrently expressed in the anterior eye. In this study, we used in vivo knockdown experiments to determine the role of PlexinD1 during vascular patterning in the anterior eye of the developing avian embryos. Knockdown of PlexinD1 in the anterior eye caused mispatterning of the vascular network in the presumptive iris, which was accompanied by lose of vascular integrity and profuse hemorrhaging in the anterior chamber. We also observed ectopic vascularization of the cornea in PlexinD1 knockdown eyes, which coincided with the formation of the limbal vasculature in controls. Finally we show that Sema3E and Sema3C transcripts are expressed in ocular tissue that is devoid of vasculature. These results indicate that PlexinD1 plays a critical role during vascular patterning in the iris and limbus, and is essential for the establishment of corneal avascularity during development. We conclude that PlexinD1 is involved in vascular response to antiangiogenic Sema3 signaling that guides the formation of the iris and limbal blood vessels by inhibiting VEGF signaling.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Córnea/irrigação sanguínea , Córnea/embriologia , Neovascularização Fisiológica/genética , Organogênese/genética , Animais , Proteínas Aviárias/biossíntese , Proteínas Aviárias/genética , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Embrião de Galinha , Hemorragia/embriologia , Hemorragia/genética , Hifema/epidemiologia , Hifema/genética , Iris/irrigação sanguínea , Iris/embriologia , Organogênese/fisiologia , Codorniz , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/genética , Semaforinas/biossíntese , Semaforinas/genética
4.
Dev Biol ; 391(2): 241-50, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24809797

RESUMO

Corneal avascularity is important for optical clarity and normal vision. However, the molecular mechanisms that prevent angioblast migration and vascularization of the developing cornea are not clear. Previously we showed that periocular angioblasts and forming ocular blood vessels avoid the presumptive cornea despite dynamic ingression of neural crest cells. In the current study, we investigate the role of Semaphorin3A (Sema3A), a cell guidance chemorepellent, on angioblast migration and corneal avascularity during development. We show that Sema3A, Vegf, and Nrp1 are expressed in the anterior eye during cornea development. Sema3A mRNA transcripts are expressed at significantly higher levels than Vegf in the lens that is positioned adjacent to the presumptive cornea. Blockade of Sema3A signaling via lens removal or injection of a synthetic Sema3A inhibitor causes ectopic migration of angioblasts into the cornea and results in its subsequent vascularization. In addition, using bead implantation, we demonstrate that exogenous Sema3A protein inhibits Vegf-induced vascularization of the cornea. In agreement with these findings, loss of Sema/Nrp1 signaling in Nrp1(Sema-) mutant mice results in ectopic angioblasts and vascularization of the embryonic mouse corneas. Altogether, our results reveal Sema3A signaling as an important cue during the establishment of corneal avascularity in both chick and mouse embryos. Our study introduces cornea development as a new model for studying the mechanisms involved in vascular patterning during embryogenesis and it also provides new insights into therapeutic potential for Sema3A in neovascular diseases.


Assuntos
Córnea/irrigação sanguínea , Cristalino/irrigação sanguínea , Neuropilina-1/genética , Semaforina-3A/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Geneticamente Modificados , Movimento Celular , Células Cultivadas , Embrião de Galinha , Córnea/embriologia , Células Endoteliais , Cristalino/embriologia , Camundongos , Neovascularização Fisiológica , Neuropilina-1/biossíntese , Codorniz/embriologia , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/genética , Semaforina-3A/antagonistas & inibidores , Semaforina-3A/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/biossíntese
5.
Dev Dyn ; 242(6): 738-51, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23444323

RESUMO

BACKGROUND: During embryonic development, endothelial precursor cells (angioblasts) migrate relatively long distances to form the primary vascular plexus. The migratory behavior of angioblasts and localization of the primitive blood vessels is tightly regulated by pro-angiogenic and anti-angiogenic factors encountered in the embryonic environment. Despite the importance of corneal avascularity to proper vision, it is not known when avascularity is established in the developing cornea and how pro- and anti-angiogenic factors regulate this process. RESULTS AND DISCUSSION: Using Tg(tie1:H2B:eYFP) transgenic quail embryos to visualize fluorescently labeled angioblasts, we show that the presumptive cornea remains avascular despite the invasion of cells from the periocular region where migratory angioblasts reside and form the primary vasculature. Semiquantitative reverse transcriptase polymerase chain reaction analysis and spatiotemporal examination of gene expression revealed that pro- and anti-angiogenic factors were expressed in patterns indicating their potential roles in angioblast guidance. CONCLUSIONS: Our findings show for the first time that chick corneal avascularity is established and maintained during development as the periocular vasculature forms. We also identify potential candidate pro- and anti-angiogenic factors that may play crucial roles during vascular patterning in the anterior eye.


Assuntos
Indutores da Angiogênese/metabolismo , Córnea/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica , Codorniz/embriologia , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/metabolismo , Córnea/irrigação sanguínea , Olho/irrigação sanguínea , Hibridização In Situ , Proteínas Luminescentes/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
bioRxiv ; 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39229219

RESUMO

Unidirectional airflow in the avian lung enables gas exchange during both inhalation and exhalation. The underlying developmental process and how it deviates from that of the bidirectional mammalian lung are poorly understood. Sampling key developmental stages with multiscale 3D imaging and single-cell transcriptomics, we delineate morphogenic, molecular, and cellular features that accommodate the unidirectional airflow in the chicken lung. Primary termini of hyper-elongated branches are eliminated via proximal-short and distal-long fusions, forming parabronchi. Neoform termini extend radially through parabronchial smooth muscle to form gas-exchanging alveoli. Supporting this radial alveologenesis, branch stalks halt their proximalization, defined by SOX9-SOX2 transition, and become SOX9 low parabronchi. Primary and secondary vascular plexi interface with primary and neoform termini, respectively. Single-cell and Stereo-seq spatial transcriptomics reveal a third, chicken-specific alveolar cell type expressing KRT14, hereby named luminal cells. Luminal, alveolar type 2, and alveolar type 1 cells sequentially occupy concentric zones radiating from the parabronchial lumen. Our study explores the evolutionary space of lung diversification and lays the foundation for functional analysis of species-specific genetic determinants.

7.
Dev Biol ; 363(1): 115-27, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22236962

RESUMO

The cornea, the most densely innervated tissue on the surface of the body, becomes innervated in a series of highly coordinated developmental events. During cornea development, chick trigeminal nerve growth cones reach the cornea margin at embryonic day (E)5, where they are initially repelled for days from E5 to E8, instead encircling the corneal periphery in a nerve ring prior to entering on E9. The molecular events coordinating growth cone guidance during cornea development are poorly understood. Here we evaluated a potential role for the Robo-Slit nerve guidance family. We found that Slits 1, 2 and 3 expression in the cornea and lens persisted during all stages of cornea innervation examined. Robo1 expression was developmentally regulated in trigeminal cell bodies, expressed robustly during nerve ring formation (E5-8), then later declining concurrent with projection of growth cones into the cornea. In this study we provide in vivo and in vitro evidence that Robo-Slit signaling guides trigeminal nerves during cornea innervation. Transient, localized inhibition of Robo-Slit signaling, by means of beads loaded with inhibitory Robo-Fc protein implanted into the developing eyefield in vivo, led to disorganized nerve ring formation and premature cornea innervation. Additionally, when trigeminal explants (source of neurons) were oriented adjacent to lens vesicles or corneas (source of repellant molecules) in organotypic tissue culture both lens and cornea tissues strongly repelled E7 trigeminal neurites, except in the presence of inhibitory Robo-Fc protein. In contrast, E10 trigeminal neurites were not as strongly repelled by cornea, and presence of Robo-Slit inhibitory protein had no effect. In full, these findings suggest that nerve repulsion from the lens and cornea during nerve ring formation is mediated by Robo-Slit signaling. Later, a shift in nerve guidance behavior occurs, in part due to molecular changes in trigeminal neurons, including Robo1 downregulation, thus allowing nerves to find the Slit-expressing cornea permissive for growth cones.


Assuntos
Córnea/metabolismo , Glicoproteínas/genética , Cápsula do Cristalino/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores Imunológicos/genética , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Embrião de Galinha , Galinhas , Córnea/embriologia , Córnea/inervação , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Cápsula do Cristalino/embriologia , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Técnicas de Cultura de Órgãos , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Tempo , Nervo Trigêmeo/citologia , Nervo Trigêmeo/embriologia , Nervo Trigêmeo/metabolismo , Proteínas Roundabout
8.
Nat Neurosci ; 11(3): 269-76, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18278043

RESUMO

Vertebrate cranial sensory ganglia, responsible for sensation of touch, taste and pain in the face and viscera, are composed of both ectodermal placode and neural crest cells. The cellular and molecular interactions allowing generation of complex ganglia remain unknown. Here, we show that proper formation of the trigeminal ganglion, the largest of the cranial ganglia, relies on reciprocal interactions between placode and neural crest cells in chick, as removal of either population resulted in severe defects. We demonstrate that ingressing placode cells express the Robo2 receptor and early migrating cranial neural crest cells express its cognate ligand Slit1. Perturbation of this receptor-ligand interaction by blocking Robo2 function or depleting either Robo2 or Slit1 using RNA interference disrupted proper ganglion formation. The resultant disorganization mimics the effects of neural crest ablation. Thus, our data reveal a novel and essential role for Robo2-Slit1 signaling in mediating neural crest-placode interactions during trigeminal gangliogenesis.


Assuntos
Movimento Celular/genética , Glicoproteínas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/embriologia , Receptores Imunológicos/metabolismo , Células-Tronco/metabolismo , Gânglio Trigeminal/embriologia , Animais , Comunicação Celular/genética , Diferenciação Celular/genética , Embrião de Galinha , Galinhas , Coturnix , Regulação para Baixo/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicoproteínas/genética , Proteínas do Tecido Nervoso/genética , Crista Neural/citologia , Crista Neural/metabolismo , Interferência de RNA , Receptores Imunológicos/genética , Células-Tronco/citologia , Gânglio Trigeminal/citologia , Gânglio Trigeminal/metabolismo , Proteínas Roundabout
9.
NPJ Regen Med ; 7(1): 36, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35879352

RESUMO

Often acute damage to the cornea initiates drastic tissue remodeling, resulting in fibrotic scarring that disrupts light transmission and precedes vision impairment. Very little is known about the factors that can mitigate fibrosis and promote scar-free cornea wound healing. We previously described transient myofibroblast differentiation during non-fibrotic repair in an embryonic cornea injury model. Here, we sought to elucidate the mechanistic regulation of myofibroblast differentiation during embryonic cornea wound healing. We found that alpha-smooth muscle actin (αSMA)-positive myofibroblasts are superficial and their presence inversely correlates with wound closure. Expression of TGFß2 and nuclear localization of pSMAD2 were elevated during myofibroblast induction. BMP3 and BMP7 were localized in the corneal epithelium and corresponded with pSMAD1/5/8 activation and absence of myofibroblasts in the healing stroma. In vitro analyses with corneal fibroblasts revealed that BMP3 inhibits the persistence of TGFß2-induced myofibroblasts by promoting disassembly of focal adhesions and αSMA fibers. This was confirmed by the expression of vinculin and pFAK. Together, these data highlight a mechanism to inhibit myofibroblast persistence during cornea wound repair.

10.
Dev Biol ; 336(2): 257-65, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19833121

RESUMO

Cranial neural crest cells migrate into the periocular region and later contribute to various ocular tissues including the cornea, ciliary body and iris. After reaching the eye, they initially pause before migrating over the lens to form the cornea. Interestingly, removal of the lens leads to premature invasion and abnormal differentiation of the cornea. In exploring the molecular mechanisms underlying this effect, we find that semaphorin3A (Sema3A) is expressed in the lens placode and epithelium continuously throughout eye development. Interestingly, neuropilin-1 (Npn-1) is expressed by periocular neural crest but down-regulated, in a manner independent of the lens, by the subpopulation that migrates into the eye and gives rise to the cornea endothelium and stroma. In contrast, Npn-1 expressing neural crest cells remain in the periocular region and contribute to the anterior uvea and ocular blood vessels. Introduction of a peptide that inhibits Sema3A/Npn-1 signaling results in premature entry of neural crest cells over the lens that phenocopies lens ablation. Furthermore, Sema3A inhibits periocular neural crest migration in vitro. Taken together, our data reveal a novel and essential role of Sema3A/Npn-1 signaling in coordinating periocular neural crest migration that is vital for proper ocular development.


Assuntos
Córnea/embriologia , Crista Neural/embriologia , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais , Animais , Embrião de Galinha , Coturnix , Imuno-Histoquímica , Hibridização In Situ
11.
Prog Mol Biol Transl Sci ; 134: 43-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26310148

RESUMO

Development of the vertebrate cornea is a multistep process that involves cellular interactions between various ectodermal-derived tissues. Bilateral interactions between the neural ectoderm-derived optic vesicles and the cranial ectoderm give rise to the presumptive corneal epithelium and other epithelia of the ocular surface. Interactions between the neural tube and the adjacent ectoderm give rise to the neural crest cells, a highly migratory and multipotent cell population. Neural crest cells migrate between the lens and presumptive corneal epithelium to form the corneal endothelium and the stromal keratocytes. The sensory nerves that abundantly innervate the corneal stroma and epithelium originate from the neural crest- and ectodermal placode-derived trigeminal ganglion. Concomitant with corneal innervation is the formation of the limbal vascular plexus and the establishment of corneal avascularity. This review summarizes historical and current research to provide an overview of the genesis of the cellular layers of the cornea, corneal innervation, and avascularity.


Assuntos
Córnea/citologia , Córnea/embriologia , Células-Tronco/citologia , Animais , Córnea/irrigação sanguínea , Córnea/inervação , Substância Própria/citologia , Substância Própria/embriologia , Desenvolvimento Embrionário , Endotélio Corneano/citologia , Endotélio Corneano/embriologia , Epitélio Corneano/citologia , Epitélio Corneano/embriologia , Humanos
12.
Methods Mol Biol ; 1189: 255-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25245699

RESUMO

The quail-chick chimera is a stable and precise labeling technique that allows tracing of definite cells and their progeny without interfering with normal development of two related avian species. This technique utilizes the transplantation of quail tissues into chick embryo or vice versa. The region of interest (graft) is removed from the donor and replaced in the stage-matched host embryo. The quail-chick chimeras can be analyzed by immunolabeling donor-derived cells with species-specific antibodies, or by differential staining of the nucleus. The use of the quail-chick chimera technique is valuable to eye development studies since its formation involves coalition of tissues from different embryonic origins: the ectoderm, neuroectoderm, and neural crest cells derived from the interaction between the ectoderm and neural ectoderm. This chapter describes the protocols for using quail-chick chimeras to identify neural crest- and ectoderm-derived components of the eye. This technique can be used in combination with molecular biology techniques in functional studies to determine the cellular and tissue interactions involved in eye development.


Assuntos
Quimera/embriologia , Coturnix/embriologia , Olho/embriologia , Animais , Embrião de Galinha , Dissecação , Ectoderma/citologia , Embrião não Mamífero/citologia , Crista Neural/citologia , Óvulo/metabolismo
13.
Invest Ophthalmol Vis Sci ; 54(9): 6334-44, 2013 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-24003085

RESUMO

PURPOSE: Wound healing in adult corneas is characterized by activation of keratocytes and extracellular matrix (ECM) synthesis that results in fibrotic scar formation and loss of transparency. Since most fetal wounds heal without scaring, we investigated the regenerative potential of wounded embryonic corneas. METHODS: On embryonic day (E) 7 chick corneas were wounded by making a linear incision traversing the epithelium and anterior stroma. Wounded corneas were collected between E7 and E18, and analyzed for apoptosis, cell proliferation, staining of ECM components, and corneal innervation. RESULTS: Substantial wound retraction was observed within 16-hours postwounding (hpw) and partial re-epithelialized by 5-days postwounding (dpw). Corneal wounds were fully re-epithelialized by 11 dpw with no visible scars. There was no difference in the number of cells undergoing apoptosis between wounded and control corneas. Cell proliferation was reduced in the wounded corneas, albeit mitotic cells in the regenerating epithelium. Staining for alpha-smooth muscle actin (α-SMA), tenascin, and fibronectin was vivid but transient at the wound site. Staining for procollagen I, perlecan, and keratan sulfate proteoglycan was reduced at the wound site. Wounded corneas were fully regenerated by 11 dpw and showed similar patterns of staining for ECM components, albeit an increase in perlecan staining. Corneal innervation was inhibited during wound healing, but regenerated corneas were innervated similar to controls. CONCLUSIONS: These data show that minimal keratocyte activation, rapid ECM reconstruction, and proper innervation occur during nonfibrotic regeneration of the embryonic cornea.


Assuntos
Córnea/embriologia , Traumatismos Oculares/patologia , Regeneração Nervosa/fisiologia , Cicatrização/fisiologia , Animais , Embrião de Galinha , Córnea/inervação , Lesões da Córnea , Modelos Animais de Doenças , Traumatismos Oculares/embriologia
14.
Stem Cells Dev ; 22(15): 2186-95, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23461574

RESUMO

The human cornea contains stem cells that can be induced to express markers consistent with multipotency in cell culture; however, there have been no studies demonstrating that human corneal keratocytes are multipotent. The objective of this study is to examine the potential of human fetal keratocytes (HFKs) to differentiate into neural crest-derived tissues when challenged in an embryonic environment. HFKs were injected bilaterally into the cranial mesenchyme adjacent to the neural tube and the periocular mesenchyme in chick embryos at embryonic days 1.5 and 3, respectively. The injected keratocytes were detected by immunofluorescence using the human cell-specific marker, HuNu. HuNu-positive keratocytes injected along the neural crest pathway were localized adjacent to HNK-1-positive migratory host neural crest cells and in the cardiac cushion mesenchyme. The HuNu-positive cells transformed into neural crest derivatives such as smooth muscle in cranial blood vessels, stromal keratocytes, and corneal endothelium. However, they failed to form neurons despite their presence in the condensing trigeminal ganglion. These results show that HFKs retain the ability to differentiate into some neural crest-derived tissues. Their ability to respond to embryonic cues and generate corneal endothelium and stromal keratocytes provides a basis for understanding the feasibility of creating specialized cells for possible use in regenerative medicine.


Assuntos
Diferenciação Celular , Ceratócitos da Córnea/fisiologia , Endotélio Corneano/citologia , Animais , Movimento Celular , Células Cultivadas , Embrião de Galinha , Córnea/citologia , Córnea/embriologia , Ceratócitos da Córnea/transplante , Vasos Coronários/citologia , Endotélio Corneano/embriologia , Feto/citologia , Humanos , Microinjeções , Crista Neural/citologia , Crânio/irrigação sanguínea , Gânglio Trigeminal/citologia
15.
Gene Expr Patterns ; 13(8): 303-10, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23727298

RESUMO

Vertebrate eye development is a complex multistep process coordinated by signals from the lens, optic cup and periocular mesenchyme. Although chemokines are increasingly being recognized as key players in cell migration, proliferation, and differentiation during embryonic development, their potential role during eye development has not been examined. In this study, we demonstrate by section in situ hybridization that CXCL12 and CXCL14 are expressed during ocular development. CXCL12 is expressed in the periocular mesenchyme, ocular blood vessels, retina, and eyelid mesenchyme, and its expression pattern is conserved between chick and mouse in most tissues. Expression of CXCL14 is localized in the ocular ectoderm, limbal epithelium, scleral papillae, eyelid mesenchyme, corneal keratocytes, hair follicles, and retina, and it was only conserved in the upper eyelid ectoderm of chick and mouse. The unique and non-overlapping patterns of CXCL12 and CXCL14 expression in ocular tissues suggest that these two chemokines may interact and have important functions in cell proliferation, differentiation and migration during eye development.


Assuntos
Proteínas Aviárias/metabolismo , Quimiocina CXCL12/metabolismo , Quimiocinas CXC/metabolismo , Olho/metabolismo , Animais , Proteínas Aviárias/genética , Quimiocina CXCL12/genética , Quimiocinas CXC/genética , Embrião de Galinha , Olho/citologia , Olho/embriologia , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos
16.
J Vis Exp ; (60)2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22349214

RESUMO

Avian embryos provide a unique platform for studying many vertebrate developmental processes, due to the easy access of the embryos within the egg. Chimeric avian embryos, in which quail donor tissue is transplanted into a chick embryo in ovo, combine the power of indelible genetic labeling of cell populations with the ease of manipulation presented by the avian embryo. Quail-chick chimeras are a classical tool for tracing migratory neural crest cells (NCCs). NCCs are a transient migratory population of cells in the embryo, which originate in the dorsal region of the developing neural tube. They undergo an epithelial to mesenchymal transition and subsequently migrate to other regions of the embryo, where they differentiate into various cell types including cartilage, melanocytes, neurons and glia. NCCs are multipotent, and their ultimate fate is influenced by 1) the region of the neural tube in which they originate along the rostro-caudal axis of the embryo, 2) signals from neighboring cells as they migrate, and 3) the microenvironment of their ultimate destination within the embryo. Tracing these cells from their point of origin at the neural tube, to their final position and fate within the embryo, provides important insight into the developmental processes that regulate patterning and organogenesis. Transplantation of complementary regions of donor neural tube (homotopic grafting) or different regions of donor neural tube (heterotopic grafting) can reveal differences in pre-specification of NCCs along the rostro-caudal axis. This technique can be further adapted to transplant a unilateral compartment of the neural tube, such that one side is derived from donor tissue, and the contralateral side remains unperturbed in the host embryo, yielding an internal control within the same sample. It can also be adapted for transplantation of brain segments in later embryos, after HH10, when the anterior neural tube has closed. Here we report techniques for generating quail-chick chimeras via neural tube transplantation, which allow for tracing of migratory NCCs derived from a discrete segment of the neural tube. Species-specific labeling of the donor-derived cells with the quail-specific QCPN antibody allows the researcher to distinguish donor and host cells at the experimental end point. This technique is straightforward, inexpensive, and has many applications, including fate-mapping, cell lineage tracing, and identifying pre-patterning events along the rostro-caudal axis. Because of the ease of access to the avian embryo, the quail-chick graft technique may be combined with other manipulations, including but not limited to lens ablation, injection of inhibitory molecules, or genetic manipulation via electroporation of expression plasmids, to identify the response of particular migratory streams of NCCs to perturbations in the embryo's developmental program. Furthermore, this grafting technique may also be used to generate other interspecific chimeric embryos such as quail-duck chimeras to study NCC contribution to craniofacial morphogenesis, or mouse-chick chimeras to combine the power of mouse genetics with the ease of manipulation of the avian embryo.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Crista Neural/citologia , Crista Neural/transplante , Quimeras de Transplante/embriologia , Transplante Heterólogo/métodos , Animais , Rastreamento de Células/métodos , Embrião de Galinha , Codorniz
17.
PLoS One ; 7(5): e37175, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615927

RESUMO

Trigeminal sensory innervation of the cornea is critical for protection and synthesis of neuropeptides required for normal vision. Little is known about axon guidance during mammalian corneal innervation. In contrast to the chick where a pericorneal nerve ring forms via Npn/Sema signaling, mouse corneal axons project directly into the presumptive cornea without initial formation of an analogous nerve ring. Here we show that during development of the mouse cornea, Npn1 is strongly expressed by the trigeminal ganglion whereas Npn2 is expressed at low levels. At the same time Sema3A and Sema3F are expressed in distinct patterns in the ocular tissues. Npn1(sema-/-) mutant corneas become precociously and aberrantly innervated by nerve bundles that project further into the corneal stroma. In contrast, stromal innervation was not affected in Npn2(-/-) mutants. The corneal epithelium was prematurely innervated in both Npn1(sema-/-) and Npn2(-/-) mutants. These defects were exacerbated in Npn1(sema-/-);Npn2(-/-) double mutants, which in addition showed ectopic innervation of the region between the optic cup and lens vesicle. Collectively, our data show that Sema3A/Npn1 and Sema3F/Npn2 signaling play distinct roles and both are required for proper innervation of the mouse cornea.


Assuntos
Substância Própria/fisiologia , Neuropilina-1/fisiologia , Neuropilina-2/fisiologia , Animais , Axônios/metabolismo , Substância Própria/metabolismo , Epitélio Corneano/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Neuropeptídeos/fisiologia , Neuropilina-1/genética , Neuropilina-1/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Transdução de Sinais , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/fisiologia
18.
Invest Ophthalmol Vis Sci ; 52(1): 30-5, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20811061

RESUMO

PURPOSE: Dense innervation of the cornea is important for maintaining its homeostasis and transparency. Although corneal nerves have been well studied in adults, little is known about mammalian corneal innervation during development. This study provides a detailed profile of nerves at various stages of mouse cornea development. METHODS: Mouse heads and corneas were collected at various stages of development including embryonic days (E)12.5 to E16.5, postnatal days (P)0, P10, three weeks after birth, and the adult. Corneas were immunostained with an anti-neuron-specific ß-tubulin antibody (TUJ1). Fluorescently labeled nerves in whole-mount tissues and sections were imaged and analyzed for their axonal projections during eye development. RESULTS: The first nerve bundles appear at the periphery of the anterior portion of the eye by E12.5. Initial projection into the stroma occurs at E13.5 without formation of a pericorneal nerve ring. Between E13.5 and E16.5, nerve bundles project directly into the periphery of the presumptive cornea stroma. They branch repeatedly as they extend toward the cornea center and epithelium. Concomitantly, nerve bundles originating from four quadrants of the eye bifurcate into smaller branches that innervate the entire stroma. The first epithelial innervation occurs at E16.5. Epithelial nerves arrange into patterns that project toward the center subsequently forming a swirl at three weeks after birth, which becomes more pronounced in adults. CONCLUSIONS: Nerve bundles that arise from four quadrants of the eye innervate the mouse cornea. The nerve bundles directly innervate the stroma without forming a pericorneal nerve ring. Radial arrangement of epithelial nerves gradually becomes centrally oriented, subsequently forming a swirl pattern.


Assuntos
Córnea/embriologia , Córnea/inervação , Desenvolvimento Embrionário/fisiologia , Nervo Oftálmico/anatomia & histologia , Nervo Oftálmico/embriologia , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Substância Própria/embriologia , Substância Própria/inervação , Epitélio Corneano/embriologia , Epitélio Corneano/inervação , Técnica Indireta de Fluorescência para Anticorpo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Nervo Oftálmico/fisiologia , Tubulina (Proteína)/metabolismo
19.
Dev Biol ; 306(2): 750-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17499699

RESUMO

The cornea, one of the most highly innervated tissues of the body, is innervated by trigeminal sensory afferents. During development, axons are initially repelled at the corneal margin, resulting in the formation of a circumferential nerve ring. The nature and source of guidance molecules that regulate this process remain a mystery. Here, we show that the lens, which immediately underlies the cornea, repels trigeminal axons in vivo and in vitro. Lens ablation results in premature, disorganized corneal innervation and disruption of the nerve ring and ventral plexus. We show that Semaphorin3A (Sema3A) is expressed in the lens epithelium and its receptor Neuropilin-1 (Npn1) is expressed in the trigeminal ganglion during cornea development. Inhibition of Sema3A signaling abrogates axon repulsion by the lens and cornea in vitro and phenocopies lens removal in vivo. These results demonstrate that lens-derived Sema3A mediates initial repulsion of trigeminal sensory axons from the cornea and is necessary for the proper formation of the nerve ring and positioning of the ventral plexus in the choroid fissure.


Assuntos
Corioide/embriologia , Córnea/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Semaforina-3A/genética , Semaforina-3A/fisiologia , Animais , Axônios/metabolismo , Embrião de Galinha , Galinhas , Hibridização In Situ , Neuropilina-1/metabolismo , Nervo Óptico/metabolismo , Peptídeos/química , Transdução de Sinais , Nervo Trigêmeo/metabolismo
20.
Dev Biol ; 288(1): 284-93, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16263107

RESUMO

Corneal keratocytes have a remarkable ability to heal the cornea throughout life. Given their developmental origin from the cranial neural crest, we asked whether this regenerative ability was related to the stem cell-like properties of their neural crest precursors. To this end, we challenged corneal stromal keratocytes by injecting them into a new environment along cranial neural crest migratory pathways. The results show that injected stromal keratocytes change their phenotype, proliferate and migrate ventrally adjacent to host neural crest cells. They then contribute to the corneal endothelial and stromal layers, the musculature of the eye, mandibular process, blood vessels and cardiac cushion tissue of the host. However, they fail to form neurons in cranial ganglia or branchial arch cartilage, illustrating that they are at least partially restricted progenitors rather than stem cells. The data show that, even at late embryonic stages, corneal keratocytes are not terminally differentiated, but maintain plasticity and multipotentiality, contributing to non-neuronal cranial neural crest derivatives.


Assuntos
Córnea/citologia , Crista Neural/citologia , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Vasos Sanguíneos/embriologia , Cartilagem/citologia , Diferenciação Celular/fisiologia , Embrião de Galinha , Corpo Ciliar/embriologia , Córnea/embriologia , Córnea/fisiologia , Coturnix/embriologia , Coração/embriologia , Iris/embriologia , Crista Neural/fisiologia , Neurônios/citologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA