Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 549: 27-33, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33662665

RESUMO

Fibroblast growth factor (FGF10)-mediated signals are essential for embryonic eyelid closure in mammals. Systemic SOX11-deficient mice are born with unclosed eyelids, suggesting a possible role of SOX11 in eyelid closure. However, the underlying mechanisms of this process remain unclear. In this study, we show that epithelial deficiency of SOX11 causes a defect in the extension of the leading edge of the eyelid, leading to failure of embryonic eyelid closure. c-Jun in the eyelid is a transcription factor downstream of FGF10 required for the extension of the leading edge of the eyelid, and c-Jun level was decreased in epithelial SOX11-deficient embryos. These results suggest that epithelial SOX11 plays an important role in embryonic eyelid closure.


Assuntos
Desenvolvimento Embrionário , Células Epiteliais/metabolismo , Pálpebras/embriologia , Pálpebras/metabolismo , Fatores de Transcrição SOXC/metabolismo , Actinas/metabolismo , Envelhecimento/patologia , Animais , Córnea/patologia , Embrião de Mamíferos/patologia , Inflamação/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fatores de Transcrição SOXC/genética
2.
Sci Signal ; 11(553)2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30352949

RESUMO

In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. We identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal-regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)-c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Our results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.


Assuntos
Receptores ErbB/fisiologia , Pálpebras/embriologia , Pálpebras/fisiologia , Lisofosfolipídeos/química , Esfingosina/análogos & derivados , Proteína ADAM10/fisiologia , Proteína ADAM17/fisiologia , Animais , Animais Geneticamente Modificados , Movimento Celular , Proteínas de Transporte de Ácido Graxo/genética , Proteínas de Transporte de Ácido Graxo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Queratinócitos/citologia , Ligantes , Fenótipo , Ratos , Transdução de Sinais , Esfingosina/química , Ativação Transcricional
3.
Exp Eye Res ; 163: 58-63, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28950938

RESUMO

The epidermal growth factor receptor (EGFR) signaling has a pivotal role in the regulation of morphogenesis during development and maintenance of homeostasis in adult eyelid and its adnexa. Studies have demonstrated that during eyelid morphogenesis the EGFR signaling pathway is responsible for keratinocyte and mesenchymal cell proliferation and migration at the eyelid tip. For meibomian gland morphogenesis, EGFR signaling activation stimulates meibomian gland epithelial cell proliferation. EGFR signaling pathway functions through multiple downstream signals such as ERK, Rho/ROCK and integrin and is regulated by a variety of upstream signals including Adam17, GPR48 and FGFR signaling. Herein we review the literature that describe the role of EGFR and its related signaling pathways in eyelid and meibomian gland morphogenesis.


Assuntos
Receptores ErbB/fisiologia , Pálpebras/embriologia , Glândulas Tarsais/embriologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Epiteliais/fisiologia , Pálpebras/fisiologia , Humanos , Glândulas Tarsais/fisiologia , Transdução de Sinais/fisiologia
4.
Ophthalmic Plast Reconstr Surg ; 32(6): 407-414, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27124372

RESUMO

PURPOSE: To review the recent data about eyelid morphogenesis, and outline a timeline for eyelid development from the very early stages during embryonic life till final maturation of the eyelid late in fetal life. METHODS: The authors extensively review major studies detailing human embryologic and fetal eyelid morphogenesis. These studies span almost a century and include some more recent cadaver studies. Numerous studies in the murine model have helped to better understand the molecular signals that govern eyelid embryogenesis. The authors summarize the current findings in molecular biology, and highlight the most significant studies in mice regarding the multiple and interacting signaling pathways involved in regulating normal eyelid morphogenesis. RESULTS: Eyelid morphogenesis involves a succession of subtle yet strictly regulated morphogenetic episodes of tissue folding, proliferation, contraction, and even migration, which may occur simultaneously or in succession. CONCLUSIONS: Understanding the extraordinary process of building eyelid tissue in embryonic life, and deciphering its underlying signaling machinery has far reaching clinical implications beyond understanding the developmental abnormalities involving the eyelids, and may pave the way for achieving scar-reducing therapies in adult mammalian wounds, or control the spread of malignancies.


Assuntos
Pálpebras/embriologia , Desenvolvimento Fetal , Humanos
5.
Artigo em Inglês | MEDLINE | ID: mdl-25419956

RESUMO

PURPOSE: To review the recent literature and describe the authors' experience with congenital upper eyelid coloboma. METHODS: In this review, we will summarize the embryologic and etiopathogenetic bases of congenital upper eyelid coloboma, and study the published clinical reports. We will also attempt to briefly shed some light on the rarer syndromic curiosities associated with upper eyelid coloboma. RESULTS: Congenital upper eyelid colobomas are one of the few nontraumatic oculoplastic emergencies that may occasionally present in the first few days of life with a corneal ulcer and may even present with impending perforation. They can present with or without corneopalpebral adhesions, may be isolated findings or a part of a larger spectrum of congenital anomalies as in the case of Fraser syndrome or Goldenhar syndrome, or could be associated with other rare curiosities that could challenge the clinician with a huge diagnostic dilemma. CONCLUSIONS: Existing literature dealing with congenital colobomas of the upper eyelid is fraught with nosologic problems, confusing etiologies, and overlapping clinical features. We attempted to clarify the salient clinical features, outline the management principles, and until a time in the not-so-distant future where advances in molecular genetic testing would help redefine the etiology and the diverse clinical spectrum of genetic diseases associated with upper eyelid colobomas, we propose a simplified classification scheme based on the relation of the coloboma to the cornea, the presence or absence of systemic features, and all the syndromic and nonsyndromic associations of congenital coloboma of the upper eyelid known today.


Assuntos
Coloboma , Pálpebras/anormalidades , Coloboma/epidemiologia , Coloboma/etiologia , Coloboma/cirurgia , Olho/embriologia , Pálpebras/embriologia , Desenvolvimento Fetal , Humanos , Procedimentos Cirúrgicos Oftalmológicos , Procedimentos de Cirurgia Plástica
6.
Dev Biol ; 395(1): 29-37, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25224220

RESUMO

Epithelial sheet movement is an essential morphogenetic process during mouse embryonic eyelid closure in which Mitogen-Activated Protein 3 Kinase 1 (MAP3K1) and c-Jun play a critical role. Here we show that MAP3K1 associates with the cytoskeleton, activates Jun N-terminal kinase (JNK) and actin polymerization, and promotes the eyelid inferior epithelial cell elongation and epithelium protrusion. Following epithelium protrusion, c-Jun begins to express and acts to promote ERK phosphorylation and migration of the protruding epithelial cells. Homozygous deletion of either gene causes defective eyelid closure, but non-allelic non-complementation does not occur between Map3k1 and c-Jun and the double heterozygotes have normal eyelid closure. Results from this study suggest that MAP3K1 and c-Jun signal through distinct temporal-spatial pathways and that productive epithelium movement for eyelid closure requires the consecutive action of MAP3K1-dependent cytoskeleton reorganization followed by c-Jun-mediated migration.


Assuntos
Epitélio/embriologia , Pálpebras/embriologia , MAP Quinase Quinase Quinase 1/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Western Blotting , Linhagem Celular , Movimento Celular/genética , Citoesqueleto/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Células HEK293 , Células HeLa , Humanos , MAP Quinase Quinase Quinase 1/genética , Células MCF-7 , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfogênese/genética , Fosforilação , Proteínas Proto-Oncogênicas c-jun/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
PLoS One ; 9(2): e87038, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498290

RESUMO

Closure of an epithelium opening is a critical morphogenetic event for development. An excellent example for this process is the transient closure of embryonic eyelid. Eyelid closure requires shape change and migration of epithelial cells at the tip of the developing eyelids, and is dictated by numerous signaling pathways. Here we evaluated gene expression in epithelial cells isolated from the tip (leading edge, LE) and inner surface epithelium (IE) of the eyelid from E15.5 mouse fetuses by laser capture microdissection (LCM). We showed that the LE and IE cells are different at E15.5, such that IE had higher expression of muscle specific genes, while LE acquired epithelium identities. Despite their distinct destinies, these cells were overall similar in expression of signaling components for the "eyelid closure pathways". However, while the LE cells had more abundant expression of Fgfr2, Erbb2, Shh, Ptch1 and 2, Smo and Gli2, and Jag1 and Notch1, the IE cells had more abundant expression of Bmp5 and Bmpr1a. In addition, the LE cells had more abundant expression of adenomatosis polyposis coli down-regulated 1 (Apcdd1), but the IE cells had high expression of Dkk2. Our results suggest that the functionally distinct LE and IE cells have also differential expression of signaling molecules that may contribute to the cell-specific responses to morphogenetic signals. The expression pattern suggests that the EGF, Shh and NOTCH pathways are preferentially active in LE cells, the BMP pathways are effective in IE cells, and the Wnt pathway may be repressed in LE and IE cells via different mechanisms.


Assuntos
Epitélio/metabolismo , Pálpebras/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais/genética , Animais , Proteínas Morfogenéticas Ósseas/genética , Fator de Crescimento Epidérmico/genética , Células Epiteliais/metabolismo , Epitélio/embriologia , Pálpebras/embriologia , Proteínas Hedgehog/genética , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Notch/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Dev Biol ; 383(2): 227-38, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24055172

RESUMO

Sproutys (Sprys) are downstream targets and negative feedback regulators of the FGF-Ras-ERK signaling pathway. Our previous studies have shown that Spry1 and Spry2, through negative modulation of FGF-ERK signaling, allow lens vesicle separation from the overlying ectoderm and regulate corneal epithelial proliferation. Here we show that Spry1 and Spry2 are necessary for eyelid closure. Murine palpebral conjunctival epithelial cells that differentiate as inner eyelids and adjacent mesenchymal cells express Spry1 and Spry2 prior to eyelid closure. Conditional deletion of both Spry1 and Spry2, but not either one alone, in the ocular surface epithelial cells result in the "EOB" (eyes open at birth) phenotype suggesting redundant roles for these proteins during eyelid closure. Spry mutant eyelids show increased proliferation of conjunctival epithelial cells with concomitant induction of FGF targets, Erm, Pea3 and Dusp6 and elevated ERK phosphorylation. Peridermal cells at the leading edge of Spry-mutant eyelids showed reduced c-Jun, but not ERK, phosphorylation, reduced F-actin polymerization and reduced motility in vitro. Spry mutant eyelids also showed disruptions in epithelial mesenchymal interactions reflected in the enhanced mesenchymal Spry1 and Spry4 expression, disaggregation of BMP4-positive mesenchymal cells and loss of Shh in the eyelid epithelium. Spry mutant eyelids also showed increased Wnt signaling and reduced expression of Foxc1 and Foxc2, two transcription factors previously shown to be necessary for eyelid closure. Collectively, our results show that conjunctival epithelial Spry1 and Spry2 redundantly promote eyelid closure by (a) stimulating ERK-independent, c-Jun-mediated peridermal migration, (b) suppressing conjunctival epithelial proliferation through FGF-ERK signaling, (c) mediating conjunctival epithelial-mesenchymal interactions and (d) maintaining expression of Foxc1 and Foxc2.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Pálpebras/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteína Morfogenética Óssea 4/metabolismo , Movimento Celular , Proliferação de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Epidérmicas , Epitélio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Pálpebras/citologia , Pálpebras/embriologia , Pálpebras/enzimologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/genética , Fosforilação , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas c-jun/metabolismo , Via de Sinalização Wnt
9.
Invest Ophthalmol Vis Sci ; 54(1): 132-40, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23211830

RESUMO

PURPOSE: During mammalian embryonic eyelid closure ADAM17 has been proposed to play a role as a transactivator of epidermal growth factor receptor (EGFR) signaling by shedding membrane bound EGFR ligands. However, ADAM17 also sheds numerous other ligands, thus implicating ADAM17 in additional molecular pathways. The goal of this study was to experimentally establish the role of ADAM17 and determine ADAM17-mediated pathways essential for the embryonic eyelid closure. METHODS: Wild-type (WT) and woe mice, carrying a hypomorphic mutation in Adam17, were evaluated using H&E and scanning electron microscopy. Expressions of ADAM17, EGFR, and the phosphorylated form EGFR-P were evaluated using immunohistochemistry. BrdU and TUNEL assays were used to evaluate cell proliferation and apoptosis, respectively. In vitro scratch assays of primary cultures were used to evaluate cell migration. Clinical and histologic analyses established if the hypermorphic Egfr(Dsk5) allele can rescue the woe embryonic eyelid closure. RESULTS: woe mice exhibited a failure to develop the leading edge of the eyelid and consequently failure of the embryonic eyelid closure. Expression of ADAM17 was identified in the eyelid epithelium in the cells of the leading edge. ADAM17 is essential for epithelial cell migration, but does not play a role in proliferation and apoptosis. EGFR was expressed in both WT and woe eyelid epithelium, but the phosphorylated EGFR-P form was detected only in WT. The Egfr(Dsk5) allele rescued woe eyelid closure defects, but also rescued woe anterior segment defects and the absence of meibomian glands. CONCLUSIONS: We provide in vivo genetic evidence that the role of ADAM17 during embryonic eyelid closure is to transactivate EGFR signaling.


Assuntos
Proteínas ADAM/metabolismo , Receptores ErbB/metabolismo , Pálpebras/embriologia , Pálpebras/metabolismo , Transdução de Sinais/fisiologia , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Morte Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Pálpebras/anormalidades , Pálpebras/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genótipo , Ligantes , Masculino , Glândulas Tarsais/citologia , Glândulas Tarsais/embriologia , Glândulas Tarsais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenótipo , Gravidez , Cultura Primária de Células
10.
Proc Natl Acad Sci U S A ; 108(42): 17349-54, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21969564

RESUMO

Developmental eyelid closure is an evolutionarily conserved morphogenetic event requiring proliferation, differentiation, cytoskeleton reorganization, and migration of epithelial cells at the tip of the developing eyelid. Many signaling events take place during eyelid closure, but how the signals converge to regulate the morphogenetic process remains an open and intriguing question. Here we show that mitogen-activated protein kinase kinase kinase 1 (MAP3K1) highly expressed in the developing eyelid epithelium, forms with c-Jun, a regulatory axis that orchestrates morphogenesis by integrating two different networks of eyelid closure signals. A TGF-α/EGFR-RhoA module initiates one of these networks by inducing c-Jun expression which, in a phosphorylation-independent manner, binds to the Map3k1 promoter and causes an increase in MAP3K1 expression. RhoA knockout in the ocular surface epithelium disturbs this network by decreasing MAP3K1 expression, and causes delayed eyelid closure in Map3k1 hemizygotes. The second network is initiated by the enzymatic activity of MAP3K1, which phosphorylates and activates a JNK-c-Jun module, leading to AP-1 transactivation and induction of its downstream genes, such as Pai-1. MAP3K1 inactivation reduces AP-1 activity and PAI-1 expression both in cells and developing eyelids. MAP3K1 is therefore the nexus of an intracrine regulatory loop connecting the TGF-α/EGFR/RhoA-c-Jun and JNK-c-Jun-AP-1 pathways in developmental eyelid closure.


Assuntos
Pálpebras/embriologia , MAP Quinase Quinase Quinase 1/metabolismo , Animais , Receptores ErbB/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , MAP Quinase Quinase Quinase 1/deficiência , MAP Quinase Quinase Quinase 1/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Modelos Biológicos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Proteínas rho de Ligação ao GTP/deficiência , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP
11.
Int J Dev Biol ; 54(10): 1473-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21136383

RESUMO

The glucocorticoid receptor (GR) plays a crucial role in epidermal morphogenesis during embryonic development, as demonstrated by analyzing genetically modified mouse models of GR gain- and loss-of-function. Eyelid formation constitutes a useful model to study epithelial development, as it requires coordinated regulation of keratinocyte proliferation, apoptosis and migration. We have analyzed this biological process in GR(-/-) embryos during ontogeny. Our data demonstrate that GR deficiency results in delayed and impaired eyelid closure, as illustrated by increased keratinocyte proliferation and apoptosis along with impaired differentiation in GR(-/-) eyelid epithelial cells. These defects are due, at least in part, to the lack of antagonism between GR and epidermal growth factor receptor (EGFR) signaling, causing sustained activation of the MAPK/AP-1 pathway and the upregulation of keratin K6 at embryonic stage E18.5. Additionally, we demonstrate that GR regulates epithelial cell migration in vitro by interfering with EGFR-mediated signaling. Overall, GR/EGFR antagonism appears as a major mechanism regulating ocular epithelial development.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Pálpebras/embriologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Animais , Apoptose , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Queratinócitos , Queratinas/genética , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfogênese , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
13.
Plast Reconstr Surg ; 124(1 Suppl): 93e-105e, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19568143

RESUMO

SUMMARY: Congenital and developmental eyelid abnormalities are among the most challenging problems encountered by the reconstructive surgeon. Eyelid abnormalities in children may present at birth as a result of abnormal embryogenesis (congenital) or they may occur at later stages as the child matures (developmental). These eyelid abnormalities include entropion, ectropion, ptosis, lid retraction, epicanthal folds, blepharophimosis, colobomas, cryptophthalmos, and canthal dystopias. Additional considerations include social factors regarding the child's self-awareness of their deformities, and specific anesthetic concerns related to their pediatric problems, which are often multisystem in nature. Current methods for evaluation and medical and surgical management are reviewed for each entity.


Assuntos
Pálpebras/anormalidades , Pálpebras/cirurgia , Entrópio/cirurgia , Pálpebras/embriologia , Humanos , Lactente
14.
Development ; 136(10): 1741-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19369394

RESUMO

There are conflicting reports about whether BMP signaling is required for eyelid closure during fetal development. This question was addressed using mice deficient in BMP or TGFbeta signaling in prospective eyelid and conjunctival epithelial cells. Genes encoding two type I BMP receptors, the type II TGFbeta receptor, two BMP- or two TGFbeta-activated R-Smads or the co-Smad Smad4 were deleted from the ocular surface ectoderm using Cre recombinase. Only mice with deletion of components of the BMP pathway had an 'eyelid open at birth' phenotype. Mice lacking Fgf10 or Fgfr2 also have open eyelids at birth. To better understand the pathways that regulate BMP expression and function during eyelid development, we localized BMPs and BMP signaling intermediates in Fgfr2 and Smad4 conditional knockout (CKO) mice. We found that Fgfr2 was required for the expression of Bmp4, the normal distribution of Shh signaling and for preserving the differentiation of the conjunctival epithelium. FGF signaling also promoted the expression of the Wnt antagonist Sfrp1 and suppressed Wnt signaling in the prospective eyelid epithelial cells, independently of BMP function. Transcripts encoding Foxc1 and Foxc2, which were previously shown to be necessary for eyelid closure, were not detectable in Smad4(CKO) animals. c-Jun, another key regulator of eyelid closure, was present and phosphorylated in eyelid periderm cells at the time of fusion, but failed to translocate to the nucleus in the absence of BMP function. Smad4(CKO) mice also showed premature differentiation of the conjunctival epithelium, conjunctival hyperplasia and the acquisition of epidermal characteristics, including formation of an ectopic row of hair follicles in place of the Meibomian glands. A second row of eyelashes is a feature of human lymphedema-distichiasis syndrome, which is associated with mutations in FOXC2.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Túnica Conjuntiva/embriologia , Túnica Conjuntiva/crescimento & desenvolvimento , Células Epiteliais/citologia , Pálpebras/embriologia , Pálpebras/crescimento & desenvolvimento , Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 4/biossíntese , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Diferenciação Celular/fisiologia , Túnica Conjuntiva/anormalidades , Túnica Conjuntiva/citologia , Células Epiteliais/metabolismo , Pálpebras/anormalidades , Fatores de Transcrição Forkhead/metabolismo , Folículo Piloso/embriologia , Folículo Piloso/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Knockout , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Proteínas Smad Reguladas por Receptor/genética , Proteína Smad4/genética , Proteína Smad4/metabolismo
15.
Invest Ophthalmol Vis Sci ; 49(10): 4245-53, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18487371

RESUMO

PURPOSE: Eyelid development is a dynamic process involving cell proliferation, differentiation, and migration regulated by a number of growth factors and cytokines. Mice deficient in the orphan G protein-coupled receptor 48 (GPR48) showed an eye open at birth (EOB) phenotype. In this study, the authors attempted to clarify the role of GPR48 in eyelid development and the molecular mechanisms leading to the EOB phenotype. METHODS: Phenotypic analysis of the eyelids of Gpr48(-/-) mice was carried out using histology and scanning electron microscopy. GPR48 expression pattern was determined using X-gal staining. In vitro scratch assay was used to determine cell motility defects in Gpr48(-)(/)(-) keratinocytes. The molecular mechanism underlying GPR48-mediated eyelid closure was explored using Western blot and immunostaining analyses. Expression levels of EGFR and its phosphorylated counterpart were examined in Gpr48(-/-) and wild-type keratinocytes and in eyelids. RESULTS: GPR48 is highly expressed in the epithelium and apical mesenchymal cells of eyelids during embryonic development. Detailed analysis revealed that Gpr48(-/-) mice exhibited delayed leading-edge extension, reduced filopodia formation, and decreased rounded periderm cell formation around eyelid margins. Keratinocytes lacking GPR48 are defective in cell proliferation and migration with reduced F-actin staining. In addition, the phosphorylation of EGFR was dramatically decreased in cultured keratinocytes and developing eyelids in the absence of GPR48. CONCLUSIONS: Inactivation of GPR48 induces the EOB phenotype by reducing epithelial cell proliferation and migration, indicating that GPR48 plays an essential role in eyelid development. Furthermore, GPR48 contributes to eyelid development through the regulation of the EGFR signaling pathway.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células , Receptores ErbB/metabolismo , Pálpebras/embriologia , Queratinócitos/citologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Western Blotting , Bromodesoxiuridina/metabolismo , Células Cultivadas , Pálpebras/metabolismo , Pálpebras/ultraestrutura , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Inativação Gênica/fisiologia , Genótipo , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Fosforilação , Transdução de Sinais/fisiologia
16.
Development ; 135(1): 23-32, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18032450

RESUMO

JNK1 and JNK2 are two ubiquitously expressed isoforms that exert redundant roles in many physiological processes, but the extent of their relative contributions to these processes has not been well characterized. We show that both JNK isoforms transmit MEK kinase 1 (MEKK1)-mediated morphogenetic signals during mouse embryonic eyelid closure. However, JNK1 and JNK2 are not synonymous, because MEKK1 is haploinsufficient for normal eyelid closure in Jnk1-null mice, but is haplosufficient in Jnk2-null mice. In the Mekk1 heterozygous background, a more efficient phosphorylation of JNK1 than JNK2 leads to differential downstream reactions, such as c-Jun phosphorylation and PAI1 expression in the developing eyelid epithelium. Differences in efficiency of phosphorylation are attributed to JNK1 Gly177 and Ser179 -- residues that are absent in JNK2 -- which promote a less ordered structural conformation. This leads to more favorable JNK phosphorylation by activin B morphogenetic signals mediated by the MEKK1-MKK4 pathway. Interestingly, Mekk1-Jnk1-Jnk2 triple hemizygotes display a partial eye-open phenotype at birth, suggesting that all three genes dose-dependently contribute to morphogenetic eyelid closure. We propose that a MEKK1-JNK1/2 axis governs the JNK activation levels to control downstream transcriptional events and eyelid morphogenesis and that reduction of upstream MEKK1 signals uncovers analogous but differential roles of JNK1 and JNK2 in a biological process.


Assuntos
Forma Celular , MAP Quinase Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Animais , Movimento Celular , Células Cultivadas , Ativação Enzimática , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Pálpebras/citologia , Pálpebras/embriologia , Pálpebras/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/enzimologia , MAP Quinase Quinase Quinase 1/genética , Camundongos , Camundongos Transgênicos , Proteína Quinase 8 Ativada por Mitógeno/deficiência , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Proteína Quinase 9 Ativada por Mitógeno/genética , Fosforilação
17.
Exp Cell Res ; 313(5): 910-20, 2007 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-17240369

RESUMO

Frem1 belongs to a family of structurally related extracellular matrix proteins of which Fras1 is the founding member. Mutations in Fras1 and Frem1 have been identified in mouse models for Fraser syndrome, which display a strikingly similar embryonic skin blistering phenotype due to impaired dermal-epidermal adhesion. Here we show that Frem1 originates from both epithelial and mesenchymal cells, in contrast to Fras1 that is exclusively derived from epithelia. However, both proteins are localized in an absolutely overlapping fashion in diverse epithelial basement membranes. At the ultrastructural level, Frem1 exhibits a clustered arrangement in the sublamina densa coinciding with fibrillar structures reminiscent of anchoring fibrils. Furthermore, in addition to its extracellular deposition, around E16, Frem1 displays an intracellular distribution in distinct epidermal cell types such as the periderm layer and basal keratinocytes. Since periderm cells are known to participate in temporary epithelial fusions like embryonic eyelid closure, defective function of Frem1 in these cells could provide a molecular explanation for the "eyes open at birth" phenotype, a feature unique for Frem1 deficient mouse mutants. Finally, we demonstrate loss of Frem1 localization in the basement membrane but not in periderm cells in the skin of Fras1(-/-) embryos. Taken together, our findings indicate that besides a cooperative function with Fras1 in embryonic basement membranes, Frem1 can also act independently in processes related to epidermal differentiation.


Assuntos
Membrana Basal/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Desenvolvimento Embrionário , Epiderme/embriologia , Epiderme/fisiologia , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular/genética , Pálpebras/embriologia , Pálpebras/fisiologia , Humanos , Mesoderma/citologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Distribuição Tecidual , Transfecção
18.
Dev Growth Differ ; 48(5): 339-46, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16759284

RESUMO

Mutant mice deficient in the fibroblast growth factor 10 (Fgf10) gene exhibit an eye-open phenotype at birth. It has previously been shown that FGF10 has a dual role in proliferation and migration during the early and later stages of eyelid development, respectively. To verify the role of FGF10 during eyelid closure, explant culture of Fgf10-null eyelid anlagen was performed, by which it was examined whether or not exogenous FGF10 could rescue the expression of activin betaB and transforming growth factor alpha, known to be required for eyelid closure. We found that the expression of these genes was markedly induced while that of Shh or Ptch1, Ptch2 was not. We also observed the distribution of filamentous actin (F-actin) after FGF10 application in the mutant eyelid explant, finding that the FGF10 protein induced F-actin accumulation. We further examined filopodia of the eyelid leading edge cells, finding the length of the filopodia was significantly reduced in the mutant. These results verify that FGF10 promotes eyelid closure through activating activin and TGFalpha-EGFR signaling.


Assuntos
Pálpebras/embriologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais , Actinas/metabolismo , Ativinas/metabolismo , Animais , Células Epiteliais , Receptores ErbB/metabolismo , Pálpebras/efeitos dos fármacos , Feminino , Feto , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/farmacologia , Proteínas Hedgehog , Masculino , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fenótipo , Pseudópodes/ultraestrutura , Transativadores/metabolismo , Fator de Crescimento Transformador alfa/metabolismo
19.
Development ; 132(19): 4317-26, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16141218

RESUMO

Heparin-binding EGF-like growth factor (HB-EGF) is a member of the EGF family of growth factors that binds to and activates the EGF receptor (EGFR) and ERBB4. Here, we show that HB-EGF-EGFR signaling is involved in eyelid development. HB-EGF expression is restricted to the tip of the leading edge of the migrating epithelium during eyelid closure in late gestation mouse embryos. Both HB-EGF null (HB(del/del)) and secretion-deficient (HB(uc/uc)) mutant embryos exhibited delayed eyelid closure, owing to slower leading edge extension and reduced actin bundle formation in migrating epithelial cells. No changes in cell proliferation were observed in these embryos. In addition, activation of EGFR and ERK was decreased in HB(del/del) eyelids. Crosses between HB(del/del) mice and waved 2 mice, a hypomorphic EGFR mutant strain, indicate that HB-EGF and EGFR interact genetically in eyelid closure. Together with our data showing that embryos treated with an EGFR-specific kinase inhibitor phenocopy HB(del/del) embryos, these data indicate that EGFR mediates HB-EGF-dependent eyelid closure. Finally, analysis of eyelid closure in TGFalpha-null mice and in HB-EGF and TGFalpha double null mice revealed that HB-EGF and TGFalpha contribute equally to and function synergistically in this process. These results indicate that soluble HB-EGF secreted from the tip of the leading edge activates the EGFR and ERK pathway, and that synergy with TGFalpha is required for leading edge extension in epithelial sheet migration during eyelid closure.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Células Epiteliais/fisiologia , Pálpebras/metabolismo , Actinas/metabolismo , Animais , Movimento Celular , Proliferação de Células , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Pálpebras/citologia , Pálpebras/embriologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout , Mutação , Transdução de Sinais , Fator de Crescimento Transformador alfa/fisiologia
20.
Development ; 132(14): 3217-30, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15958512

RESUMO

The development of the eyelid requires coordinated cellular processes of proliferation, cell shape changes, migration and cell death. Mutant mice deficient in the fibroblast growth factor 10 (Fgf10) gene exhibit open-eyelids at birth. To elucidate the roles of FGF10 during eyelid formation, we examined the expression pattern of Fgf10 during eyelid formation and the phenotype of Fgf10-null eyelids in detail. Fgf10 is expressed by mesenchymal cells just beneath the protruding epidermal cells of the nascent eyelid. However, Fgf10-null epithelial cells running though the eyelid groove do not exhibit typical cuboid shape or sufficient proliferation. Furthermore, peridermal clumps are not maintained on the eyelid leading edge, and epithelial extension does not occur. At the cellular level, the accumulation of actin fibers is not observed in the mutant epithelial leading edge. The expression of activin/inhibin betaB (ActbetaB/Inhbb) and transforming growth factor alpha (Tgfa), previously reported to be crucial for eyelid development, is down-regulated in the mutant leading edge, while the onset of sonic hedgehog (Shh) expression is delayed on the mutant eyelid margin. Explant cultures of mouse eyelid primordia shows that the open-eyelid phenotype of the mutant is reduced by exogenous FGF10 protein, and that the expression of ActbetaB and Tgfa is ectopically induced in the thickened eyelid epithelium by the FGF10 protein. These results indicate a dual role of FGF10 in mouse eyelid development, for both proliferation and coordinated migration of eyelid epithelial cells by reorganization of the cytoskeleton, through the regulation of activin, TGFalpha and SHH signaling.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células , Células Epiteliais/metabolismo , Pálpebras/embriologia , Fatores de Crescimento de Fibroblastos/fisiologia , Actinas/metabolismo , Animais , Movimento Celular/genética , Fator 10 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/genética , Proteínas Hedgehog , Queratinócitos/citologia , Queratinócitos/fisiologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Pseudópodes/fisiologia , Transativadores/metabolismo , Fator de Crescimento Transformador alfa/biossíntese , Fator de Crescimento Transformador alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA