Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dis Model Mech ; 13(8)2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32571845

RESUMO

Wnt/ß-catenin signaling has an essential role in eye development. Faulty regulation of this pathway results in ocular malformations, owing to defects in cell-fate determination and differentiation. Herein, we show that disruption of Maz, the gene encoding Myc-associated zinc-finger transcription factor, produces developmental eye defects in mice and humans. Expression of key genes involved in the Wnt cascade, Sfrp2, Wnt2b and Fzd4, was significantly increased in mice with targeted inactivation of Maz, resulting in abnormal peripheral eye formation with reduced proliferation of the progenitor cells in the region. Paradoxically, the Wnt reporter TCF-Lef1 displayed a significant downregulation in Maz-deficient eyes. Molecular analysis indicates that Maz is necessary for the activation of the Wnt/ß-catenin pathway and participates in the network controlling ciliary margin patterning. Copy-number variations and single-nucleotide variants of MAZ were identified in humans that result in abnormal ocular development. The data support MAZ as a key contributor to the eye comorbidities associated with chromosome 16p11.2 copy-number variants and as a transcriptional regulator of ocular development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Anormalidades do Olho/metabolismo , Olho/metabolismo , Fatores de Transcrição/metabolismo , Adolescente , Adulto , Animais , Apoptose , Proliferação de Células , Pré-Escolar , Variações do Número de Cópias de DNA , Proteínas de Ligação a DNA/genética , Olho/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Humanos , Lactente , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Transcrição/genética , Via de Sinalização Wnt , Adulto Jovem
2.
J Clin Invest ; 126(3): 948-61, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26854927

RESUMO

The ascending thoracic aorta is designed to withstand biomechanical forces from pulsatile blood. Thoracic aortic aneurysms and acute aortic dissections (TAADs) occur as a result of genetically triggered defects in aortic structure and a dysfunctional response to these forces. Here, we describe mutations in the forkhead transcription factor FOXE3 that predispose mutation-bearing individuals to TAAD. We performed exome sequencing of a large family with multiple members with TAADs and identified a rare variant in FOXE3 with an altered amino acid in the DNA-binding domain (p.Asp153His) that segregated with disease in this family. Additional pathogenic FOXE3 variants were identified in unrelated TAAD families. In mice, Foxe3 deficiency reduced smooth muscle cell (SMC) density and impaired SMC differentiation in the ascending aorta. Foxe3 expression was induced in aortic SMCs after transverse aortic constriction, and Foxe3 deficiency increased SMC apoptosis and ascending aortic rupture with increased aortic pressure. These phenotypes were rescued by inhibiting p53 activity, either by administration of a p53 inhibitor (pifithrin-α), or by crossing Foxe3-/- mice with p53-/- mice. Our data demonstrate that FOXE3 mutations lead to a reduced number of aortic SMCs during development and increased SMC apoptosis in the ascending aorta in response to increased biomechanical forces, thus defining an additional molecular pathway that leads to familial thoracic aortic disease.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Fatores de Transcrição Forkhead/genética , Adulto , Dissecção Aórtica/metabolismo , Dissecção Aórtica/patologia , Animais , Aorta/metabolismo , Aorta/patologia , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/patologia , Apoptose , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Mutação de Sentido Incorreto , Miócitos de Músculo Liso/fisiologia , Linhagem , Proteína Supressora de Tumor p53/genética , Remodelação Vascular , Peixe-Zebra
3.
Am J Pathol ; 185(1): 197-213, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25451153

RESUMO

Wnt glycoproteins control key processes during development and disease by activating various downstream pathways. Wnt secretion requires post-translational modification mediated by the O-acyltransferase encoded by the Drosophila porcupine homolog gene (PORCN). In humans, PORCN mutations cause focal dermal hypoplasia (FDH, or Goltz syndrome), an X-linked dominant multisystem birth defect that is frequently accompanied by ocular abnormalities such as coloboma, microphthalmia, or even anophthalmia. Although genetic ablation of Porcn in mouse has provided insight into the etiology of defects caused by ectomesodermal dysplasia in FDH, the requirement for Porcn and the actual Wnt ligands during eye development have been unknown. In this study, Porcn hemizygosity occasionally caused ocular defects reminiscent of FDH. Conditional inactivation of Porcn in periocular mesenchyme led to defects in mid- and hindbrain and in craniofacial development, but was insufficient to cause ocular abnormalities. However, a combination of conditional Porcn depletion in optic vesicle neuroectoderm, lens, and neural crest-derived periocular mesenchyme induced severe eye abnormalities with high penetrance. In particular, we observed coloboma, transdifferentiation of the dorsal and ventral retinal pigment epithelium, defective optic cup periphery, and closure defects of the eyelid, as well as defective corneal morphogenesis. Thus, Porcn is required in both extraocular and neuroectodermal tissues to regulate distinct Wnt-dependent processes during morphogenesis of the posterior and anterior segments of the eye.


Assuntos
Olho/embriologia , Hipoplasia Dérmica Focal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Aciltransferases , Alelos , Animais , Modelos Animais de Doenças , Olho/metabolismo , Feminino , Genótipo , Glicoproteínas/metabolismo , Hemizigoto , Hibridização In Situ , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Recombinação Genética , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/metabolismo , Proteínas Wnt/metabolismo
4.
J Neurosci ; 33(45): 17847-62, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24198374

RESUMO

The present study examined the consequences of eliminating horizontal cells from the outer retina during embryogenesis upon the organization and assembly of the outer plexiform layer (OPL). Retinal horizontal cells exhibit a migration defect in Lim1-conditional knock-out (Lim1-CKO) mice and become mispositioned in the inner retina before birth, redirecting their dendrites into the inner plexiform layer. The resultant (mature) OPL, developing in the absence of horizontal cells, shows a retraction of rod spherules into the outer nuclear layer and a sprouting of rod bipolar cell dendrites to reach ectopic ribbon-protein puncta. Cone pedicles and the dendrites of type 7 cone bipolar cells retain their characteristic stratification and colocalization within the collapsed OPL, although both are atrophic and the spatial distribution of the pedicles is disrupted. Developmental analysis of Lim1-CKO retina reveals that components of the rod and cone pathways initially co-assemble within their normal strata in the OPL, indicating that horizontal cells are not required for the correct targeting of photoreceptor terminals or bipolar cell dendrites. As the rod spherules begin to retract during the second postnatal week, rod bipolar cells initially show no signs of ectopic growth, sprouting only subsequently and continuing to do so well after the eighth postnatal week. These results demonstrate the critical yet distinctive roles for horizontal cells on the rod and cone pathways and highlight a unique and as-yet-unrecognized maintenance function of an inhibitory interneuron that is not required for the initial targeting and co-stratification of other components in the circuit.


Assuntos
Plasticidade Neuronal/fisiologia , Células Horizontais da Retina/fisiologia , Neurônios Retinianos/fisiologia , Vias Visuais/crescimento & desenvolvimento , Animais , Feminino , Camundongos , Camundongos Transgênicos , Sinapses/metabolismo , Vias Visuais/fisiologia
5.
Development ; 140(6): 1330-41, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23406904

RESUMO

Dysfunction or death of photoreceptors is the primary cause of vision loss in retinal and macular degenerative diseases. As photoreceptors have an intimate relationship with the retinal pigment epithelium (RPE) for exchange of macromolecules, removal of shed membrane discs and retinoid recycling, an improved understanding of the development of the photoreceptor-RPE complex will allow better design of gene- and cell-based therapies. To explore the epigenetic contribution to retinal development we generated conditional knockout alleles of DNA methyltransferase 1 (Dnmt1) in mice. Conditional Dnmt1 knockdown in early eye development mediated by Rx-Cre did not produce lamination or cell fate defects, except in cones; however, the photoreceptors completely lacked outer segments despite near normal expression of phototransduction and cilia genes. We also identified disruption of RPE morphology and polarization as early as E15.5. Defects in outer segment biogenesis were evident with Dnmt1 exon excision only in RPE, but not when excision was directed exclusively to photoreceptors. We detected a reduction in DNA methylation of LINE1 elements (a measure of global DNA methylation) in developing mutant RPE as compared with neural retina, and of Tuba3a, which exhibited dramatically increased expression in mutant retina. These results demonstrate a unique function of DNMT1-mediated DNA methylation in controlling RPE apicobasal polarity and neural retina differentiation. We also establish a model to study the epigenetic mechanisms and signaling pathways that guide the modulation of photoreceptor outer segment morphogenesis by RPE during retinal development and disease.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , DNA (Citosina-5-)-Metiltransferases/genética , Morfogênese/genética , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Epitélio Pigmentado da Retina/fisiologia , Animais , Permeabilidade da Membrana Celular/genética , Polaridade Celular/genética , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Morfogênese/fisiologia , Especificidade de Órgãos/genética , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/crescimento & desenvolvimento , Epitélio Pigmentado da Retina/metabolismo , Transcriptoma
6.
J Neurosci ; 33(1): 259-72, 2013 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-23283339

RESUMO

The brain plays a central role in controlling energy, glucose, and lipid homeostasis, with specialized neurons within nuclei of the mediobasal hypothalamus, namely the arcuate (ARC) and ventromedial (VMH), tasked with proper signal integration. Exactly how the exquisite cytoarchitecture and underlying circuitry becomes established within these nuclei remains largely unknown, in part because hypothalamic developmental programs are just beginning to be elucidated. Here, we demonstrate that the Retina and anterior neural fold homeobox (Rax) gene plays a key role in establishing ARC and VMH nuclei in mice. First, we show that Rax is expressed in ARC and VMH progenitors throughout development, consistent with genetic fate mapping studies demonstrating that Rax+ lineages give rise to VMH neurons. Second, the conditional ablation of Rax in a subset of VMH progenitors using a Shh::Cre driver leads to a fate switch from a VMH neuronal phenotype to a hypothalamic but non-VMH identity, suggesting that Rax is a selector gene for VMH cellular fates. Finally, the broader elimination of Rax throughout ARC/VMH progenitors using Six3::Cre leads to a severe loss of both VMH and ARC cellular phenotypes, demonstrating a role for Rax in both VMH and ARC fate specification. Combined, our study illustrates that Rax is required in ARC/VMH progenitors to specify neuronal phenotypes within this hypothalamic brain region. Rax thus provides a molecular entry point for further study of the ontology and establishment of hypothalamic feeding circuits.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipotálamo Médio/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem da Célula , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Hipotálamo Médio/embriologia , Hipotálamo Médio/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Fatores de Transcrição/genética
7.
Hum Mol Genet ; 21(12): 2663-76, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22398208

RESUMO

Mutation of the polarity gene Crumbs homolog 1 (CRB1) is responsible for >10% of Leber congenital amaurosis (LCA) cases worldwide; LCA is characterized by early-onset degenerative retinal dystrophy. The role of CRB1 in LCA8 pathogenesis remains elusive since Crb1 mouse mutants, including a null allele, have failed to mimic the early-onset of LCA, most likely due to functional compensation by closely related genes encoding Crb2 and Crb3. Crb proteins form an evolutionarily conserved, apical polarity complex with the scaffolding protein associated with lin-seven 1 (Pals1), also known as MAGUK p55 subfamily member 5 (MPP5). Pals1 and Crbs are functionally inter-dependent in establishing and maintaining epithelial polarity. Pals1 is a single gene in the mouse and human genomes; therefore, we ablated Pals1 to establish a mouse genetic model mimicking human LCA. In our study, the deletion of Pals1 leads to the disruption of the apical localization of Crb proteins in retinal progenitors and the adult retina, validating their mutual interaction. Remarkably, the Pals1 mutant mouse exhibits the critical features of LCA such as early visual impairment as assessed by electroretinogram, disorganization of lamination and apical junctions and retinal degeneration. Our data uncover the indispensible role of Pals1 in retinal development, likely involving the maintenance of retinal polarity and survival of retinal neurons, thus providing the basis for the pathologic mechanisms of LCA8.


Assuntos
Amaurose Congênita de Leber/metabolismo , Proteínas de Membrana/metabolismo , Núcleosídeo-Fosfato Quinase/metabolismo , Retina/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Eletrorretinografia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica , Hibridização In Situ , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/patologia , Masculino , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Proteínas do Tecido Nervoso/metabolismo , Núcleosídeo-Fosfato Quinase/genética , Retina/embriologia , Retina/crescimento & desenvolvimento , Células-Tronco/patologia , Células-Tronco/ultraestrutura , Acuidade Visual
8.
J Comp Neurol ; 520(7): 1562-83, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22102330

RESUMO

The development of topographic maps of the sensory periphery is sensitive to the disruption of adenylate cyclase 1 (AC1) signaling. AC1 catalyzes the production of cAMP in a Ca2+/calmodulin-dependent manner, and AC1 mutant mice (AC1−/−) have disordered visual and somatotopic maps. However, the broad expression of AC1 in the brain and the promiscuous nature of cAMP signaling have frustrated attempts to determine the underlying mechanism of AC1-dependent map development. In the mammalian visual system, the initial coarse targeting of retinal ganglion cell (RGC) projections to the superior colliculus (SC) and lateral geniculate nucleus (LGN) is guided by molecular cues, and the subsequent refinement of these crude projections occurs via an activity-dependent process that depends on spontaneous retinal waves. Here, we show that AC1−/− mice have normal retinal waves but disrupted map refinement. We demonstrate that AC1 is required for the emergence of dense and focused termination zones and elimination of inaccurately targeted collaterals at the level of individual retinofugal arbors. Conditional deletion of AC1 in the retina recapitulates map defects, indicating that the locus of map disruptions in the SC and dorsal LGN of AC1−/− mice is presynaptic. Finally, map defects in mice without AC1 and disrupted retinal waves (AC1−/−;ß2−/− double KO mice) are no worse than those in mice lacking only ß2−/−, but loss of AC1 occludes map recovery in ß2−/− mice during the second postnatal week. These results suggest that AC1 in RGC axons mediates the development of retinotopy and eye-specific segregation in the SC and dorsal LGN.


Assuntos
Adenilil Ciclases/metabolismo , Mapeamento Encefálico/métodos , Células Ganglionares da Retina/metabolismo , Vias Visuais/metabolismo , Animais , Eletrofisiologia , Eletroporação , Corpos Geniculados/citologia , Corpos Geniculados/crescimento & desenvolvimento , Corpos Geniculados/metabolismo , Imageamento Tridimensional/métodos , Camundongos , Camundongos Knockout , Camundongos Mutantes , Células Ganglionares da Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Colículos Superiores/citologia , Colículos Superiores/crescimento & desenvolvimento , Colículos Superiores/metabolismo , Vias Visuais/citologia
9.
J Neurosci ; 31(47): 17230-41, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22114289

RESUMO

The membrane-associated palmitoylated protein 5 (MPP5 or PALS1) is thought to organize intracellular PALS1-CRB-MUPP1 protein scaffolds in the retina that are involved in maintenance of photoreceptor-Müller glia cell adhesion. In humans, the Crumbs homolog 1 (CRB1) gene is mutated in progressive types of autosomal recessive retinitis pigmentosa and Leber congenital amaurosis. However, there is no clear genotype-phenotype correlation for CRB1 mutations, which suggests that other components of the CRB complex may influence the severity of retinal disease. Therefore, to understand the physiological role of the Crumbs complex proteins, especially PALS1, we generated and analyzed conditional knockdown mice for Pals1. Small irregularly shaped spots were detected throughout the PALS1 deficient retina by confocal scanning laser ophthalmoscopy and spectral domain optical coherence tomography. The electroretinography a- and b-wave was severely attenuated in the aged mutant retinas, suggesting progressive degeneration of photoreceptors. The histological analysis showed abnormal retinal pigment epithelium structure, ectopic photoreceptor nuclei in the subretinal space, an irregular outer limiting membrane, half rosettes of photoreceptors in the outer plexiform layer, and a thinner photoreceptor synaptic layer suggesting improper photoreceptor cell layering during retinal development. The PALS1 deficient retinas showed reduced levels of Crumbs complex proteins adjacent to adherens junctions, upregulation of glial fibrillary acidic protein indicative of gliosis, and persisting programmed cell death after retinal maturation. The phenotype suggests important functions of PALS1 in the retinal pigment epithelium in addition to the neural retina.


Assuntos
Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Núcleosídeo-Fosfato Quinase/deficiência , Núcleosídeo-Fosfato Quinase/genética , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura , Animais , Feminino , Masculino , Marmota , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/ultraestrutura , Oftalmoscopia , Retina/metabolismo , Retina/ultraestrutura , Tomografia de Coerência Óptica
10.
J Clin Invest ; 120(10): 3568-77, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20890044

RESUMO

Retinal degeneration causes vision impairment and blindness in humans. If one day we are to harness the potential of stem cell-based cell replacement therapies to treat these conditions, it is imperative that we better understand normal retina development. Currently, the genes and mechanisms that regulate the specification of the neuroretina during vertebrate eye development remain unknown. Here, we identify sine oculis-related homeobox 3 (Six3) as a crucial player in this process in mice. In Six3 conditional-mutant mouse embryos, specification of the neuroretina was abrogated, but that of the retinal pigmented epithelium was normal. Conditional deletion of Six3 did not affect the initial development of the optic vesicle but did arrest subsequent neuroretina specification. Ectopic rostral expansion of Wnt8b expression was the major response to Six3 deletion and the leading cause for the specific lack of neuroretina, as ectopic Wnt8b expression in transgenic embryos was sufficient to suppress neuroretina specification. Using chromatin immunoprecipitation assays, we identified Six3-responsive elements in the Wnt8b locus and demonstrated that Six3 directly repressed Wnt8b expression in vivo. Our findings provide a molecular framework to the program leading to neuroretina differentiation and may be relevant for the development of novel strategies aimed at characterizing and eventually treating different abnormalities in eye formation.


Assuntos
Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Placa Neural/embriologia , Retina/embriologia , Proteínas Wnt/antagonistas & inibidores , Animais , Camundongos , Camundongos Transgênicos , Fatores de Transcrição SOXB1/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Proteína Homeobox SIX3
11.
J Clin Invest ; 120(7): 2474-85, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20530870

RESUMO

The majority of human skeletal dysplasias are caused by dysregulation of growth plate homeostasis. As TGF-beta signaling is a critical determinant of growth plate homeostasis, skeletal dysplasias are often associated with dysregulation of this pathway. The context-dependent action of TFG-beta signaling is tightly controlled by numerous mechanisms at the extracellular level and downstream of ligand-receptor interactions. However, TGF-beta is synthesized as an inactive precursor that is cleaved to become mature in the Golgi apparatus, and the regulation of this posttranslational intracellular processing and trafficking is much less defined. Here, we report that a cysteine-rich protein, E-selectin ligand-1 (ESL-1), acts as a negative regulator of TGF-beta production by binding TGF-beta precursors in the Golgi apparatus in a cell-autonomous fashion, inhibiting their maturation. Furthermore, ESL-1 inhibited the processing of proTGF-beta by a furin-like protease, leading to reduced secretion of mature TGF-beta by primary mouse chondrocytes and HEK293 cells. In vivo loss of Esl1 in mice led to increased TGF-beta/SMAD signaling in the growth plate that was associated with reduced chondrocyte proliferation and delayed terminal differentiation. Gain-of-function and rescue studies of the Xenopus ESL-1 ortholog in the context of early embryogenesis showed that this regulation of TGF-beta/Nodal signaling was evolutionarily conserved. This study identifies what we believe to be a novel intracellular mechanism for regulating TGF-beta during skeletal development and homeostasis.


Assuntos
Condrócitos/metabolismo , Lâmina de Crescimento/metabolismo , Homeostase , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Diferenciação Celular/fisiologia , Condrócitos/citologia , Citoplasma/metabolismo , Selectina E/metabolismo , Furina/metabolismo , Lâmina de Crescimento/citologia , Ligantes , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores de Fatores de Crescimento de Fibroblastos , Selectinas/metabolismo , Sialoglicoproteínas , Transdução de Sinais/fisiologia , Xenopus laevis
12.
Int J Dev Biol ; 54(5): 919-24, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20336609

RESUMO

FoxF genes are essential for visceral mesoderm development from Drosophila to human. However, part of the difficulty of studying the visceral mesoderm is its relative inaccessibility during early development. Owing to its external development Xenopus laevis presents considerable advantages for the study of visceral mesoderm formation, yet FoxF2 has not been identified in this system. Here, we describe the cloning and expression pattern of XFoxF2 during embryonic development, metamorphosis and adulthood, and compare and contrast it to the expression of FoxF1 in Xenopus laevis and FoxF2 in mouse.


Assuntos
Fatores de Transcrição Forkhead/genética , Trato Gastrointestinal/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Xenopus/genética , Xenopus laevis/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/crescimento & desenvolvimento , Hibridização In Situ , Metamorfose Biológica/genética , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Xenopus laevis/embriologia , Xenopus laevis/crescimento & desenvolvimento
13.
Development ; 137(5): 725-34, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20147377

RESUMO

Transcriptional networks, which are initiated by secreted proteins, cooperate with each other to orchestrate eye development. The establishment of dorsal/ventral polarity, especially dorsal specification in the optic vesicle, is poorly understood at a molecular and cellular level. Here, we show that COUP-TFI (Nr2f1) and COUP-TFII (Nr2f2) are highly expressed in the progenitor cells in the developing murine eye. Phenotype analysis of COUP-TFI and COUP-TFII single-gene conditional knockout mouse models suggests that COUP-TFs compensate for each other to maintain morphogenesis of the eye. However, in eye-specific COUP-TFI/TFII double-knockout mice, progenitor cells at the dorso-distal optic vesicle fail to differentiate appropriately, causing the retinal pigmented epithelium cells to adopt a neural retina fate and abnormal differentiation of the dorsal optic stalk; the development of proximo-ventral identities, neural retina and ventral optic stalk is also compromised. These cellular defects in turn lead to congenital ocular colobomata and microphthalmia. Immunohistochemical and in situ hybridization assays reveal that the expression of several regulatory genes essential for early optic vesicle development, including Pax6, Otx2, Mitf, Pax2 and Vax1/2, is altered in the corresponding compartments of the mutant eye. Using ChIP assay, siRNA treatment and transient transfection in ARPE-19 cells in vitro, we demonstrate that Pax6 and Otx2 are directly regulated by COUP-TFs. Taken together, our findings reveal novel and distinct cell-intrinsic mechanisms mediated by COUP-TF genes to direct the specification and differentiation of progenitor cells, and that COUP-TFs are crucial for dorsalization of the eye.


Assuntos
Fatores de Transcrição COUP/fisiologia , Proteínas do Olho/genética , Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética , Animais , Fatores de Transcrição COUP/genética , Células Cultivadas , Coloboma/embriologia , Coloboma/genética , Embrião de Mamíferos/metabolismo , Olho/metabolismo , Anormalidades do Olho/embriologia , Anormalidades do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Microftalmia/embriologia , Microftalmia/genética , Modelos Biológicos , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retina/embriologia , Retina/metabolismo , Vias Visuais/embriologia
14.
Dev Dyn ; 238(9): 2193-201, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19334279

RESUMO

The transcription factor Pitx3 is critical for lens formation. Deletions in the promoter of this gene cause abnormal lens development in the aphakia (ak) mouse mutant, which has only rudimentary lenses. In this study, we investigated the role of Pitx3 in lens development and differentiation. We found that reduced expression of Pitx3 leads to changes in the proliferation, differentiation and survival of lens cells. The genetic interactions between Pitx3 and Foxe3 were investigated, as these two transcription factors are expressed at the same time in lens development and their absence has similar consequences for lens development. We found no evidence that these two genes genetically interact. In general, our study shows that the abnormal phenotype of the ak lenses is not due to just one molecular pathway, rather in the absence of Pitx3 expression multiple aspects of lens development are disrupted.


Assuntos
Proteínas de Homeodomínio/fisiologia , Cristalino/embriologia , Cristalino/metabolismo , Fatores de Transcrição/fisiologia , Animais , Afacia/embriologia , Afacia/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Mutantes , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Gastroenterology ; 136(7): 2074-91, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19303014

RESUMO

The gastrointestinal (GI) tract forms from the endoderm (which gives rise to the epithelium) and the mesoderm (which develops into the smooth muscle layer, the mesenchyme, and numerous other cell types). Much of what is known of GI development has been learned from studies of the endoderm and its derivatives, because of the importance of epithelial biology in understanding and treating human diseases. Although the necessity of epithelial-mesenchymal cross talk for GI development is uncontested, the role of the mesoderm remains comparatively less well understood. The transformation of the visceral mesoderm during development is remarkable; it differentiates from a very thin layer of cells into a complex tissue comprising smooth muscle cells, myofibroblasts, neurons, immune cells, endothelial cells, lymphatics, and extracellular matrix molecules, all contributing to the form and function of the digestive system. Understanding the molecular processes that govern the development of these cell types and elucidating their respective contribution to GI patterning could offer insight into the mechanisms that regulate cell fate decisions in the intestine, which has the unique property of rapid cell renewal for the maintenance of epithelial integrity. In reviewing evidence from both mammalian and nonmammalian models, we reveal the important role of the visceral mesoderm in the ontogeny of the GI tract.


Assuntos
Desenvolvimento Embrionário/fisiologia , Trato Gastrointestinal/embriologia , Mesoderma/embriologia , Animais , Embrião de Galinha , Desenvolvimento Fetal , Trato Gastrointestinal/fisiologia , Intestino Grosso/embriologia , Intestino Delgado/embriologia , Mesoderma/fisiologia , Camundongos , Modelos Animais , Especificidade da Espécie , Estômago/embriologia , Xenopus
16.
PLoS One ; 4(2): e4513, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19229337

RESUMO

Rx is a paired-like homeobox gene that is required for vertebrate eye formation. Mice lacking Rx function do not develop eyes or the posterior pituitary. To determine whether Rx is required cell autonomously in these tissues, we generated embryonic chimeras consisting of wild type and Rx-/- cells. We found that in the eye, Rx-deficient cells cannot participate in the formation of the neuroretina, retina pigment epithelium and the distal part of the optic stalk. In addition, in the ventral forebrain, Rx function is required cell autonomously for the formation of the posterior pituitary. Interestingly, Rx-/- and wild type cells segregate before the morphogenesis of these two tissues begins. Our observations suggest that Rx function is not only required for the morphogenesis of the retina and posterior pituitary, but also prior to morphogenesis, for the sorting out of cells to form distinct fields of retinal/pituitary cells.


Assuntos
Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Morfogênese , Neuro-Hipófise/crescimento & desenvolvimento , Retina/crescimento & desenvolvimento , Animais , Movimento Celular , Quimera , Embrião de Mamíferos , Camundongos , Camundongos Knockout , Neuro-Hipófise/embriologia , Retina/embriologia
17.
Int J Dev Biol ; 52(8): 1123-33, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18956346

RESUMO

The complement system is the central component of innate immunity and an important player in the adaptive immunity of vertebrates. We analyzed the expression patterns of several key members of the complement cascade during Xenopus development. We found extensive expression of these molecules already during gastrula/early neurula stage. Remarkably, several genes also showed an organ-specific expression pattern during early organogenesis. Early expression is notable for two different expression patterns in the neuroectoderm. In one group, there is early strong neural plate and neural precursor expression. This is the case of properdin, C1qA, C3 and C9. The second pattern, seen with C1qR and C6, is noteworthy for its expression at the periphery of the neural plate, in the presumptive neural crest. Two genes stand out for their predominantly mesodermal expression. C3aR, the message for the cognate receptor for C3 in the complement cascade, is expressed at the same time as C3, but in a complementary, reciprocal pattern in the mesoderm. C1qA expression also predominates in somites, pronephros, visceral mesoderm and ventral blood islands. Finally, several genes are characterized by later expression in developing organs. C1qR displays a reticular pattern consistent with expression in the developing vasculature. The late expression of C1qA and C3bC4b is strongest in the pronephros. Finally, the expression of properdin in the hindbrain and in the developing lens are novel findings. The expression patterns of these molecules suggest that these components of the complement system may have in Xenopus a so far undefined developmental role.


Assuntos
Padronização Corporal/genética , Padronização Corporal/imunologia , Proteínas do Sistema Complemento/genética , Xenopus laevis/embriologia , Xenopus laevis/imunologia , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/imunologia , Complemento C1q/genética , Complemento C3/genética , Complemento C9/genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Cristalino/embriologia , Cristalino/imunologia , Mesoderma/embriologia , Mesoderma/imunologia , Tubo Neural/embriologia , Tubo Neural/imunologia , Organogênese/genética , Organogênese/imunologia , Properdina/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/imunologia , Xenopus laevis/genética
18.
J Biol Chem ; 283(44): 30015-24, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18713731

RESUMO

The dynamics of G protein-mediated signal transduction depend on the two-dimensional diffusion of membrane-bound G proteins and receptors, which has been suggested to be rate-limiting for vertebrate phototransduction, a highly amplified G protein-coupled signaling pathway. Using fluorescence recovery after photobleaching (FRAP), we measured the diffusion of the G protein transducin alpha-subunit (Galpha(t)) and the G protein-coupled receptor rhodopsin on disk membranes of living rod photoreceptors from transgenic Xenopus laevis. Treatment with either methyl-beta-cyclodextrin or filipin III to disrupt cholesterol-containing lipid microdomains dramatically accelerated diffusion of Galpha(t) in its GTP-bound state and of the rhodopsin-Galphabetagamma(t) complex but not of rhodopsin or inactive GDP-bound Galphabetagamma. These results imply an activity-dependent sequestration of G proteins into cholesterol-dependent lipid microdomains, which limits diffusion and exclude the majority of free rhodopsin and the free G protein heterotrimer. Our data offer a novel demonstration of lipid microdomains in the internal membranes of living sensory neurons.


Assuntos
Proteínas de Ligação ao GTP/química , Lipídeos/química , Microdomínios da Membrana/química , Animais , Colesterol/metabolismo , Densitometria , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/química , Insetos , Microscopia de Fluorescência , Neurônios/metabolismo , Células Fotorreceptoras/metabolismo , Estrutura Terciária de Proteína , Rodopsina/química , Transdução de Sinais , Xenopus laevis
19.
Dev Biol ; 322(1): 56-64, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18675797

RESUMO

Eye development is a complex process that involves the formation of the retina and the lens, collectively called the eyeball, as well as the formation of auxiliary eye structures such as the eyelid, lacrimal gland, cornea and conjunctiva. The developmental requirements for the formation of each individual structure are only partially understood. We have shown previously that the homeobox-containing gene Rx is a key component in eye formation, as retinal structures do not develop and retina-specific gene expression is not observed in Rx-deficient mice. In addition, Rx-/- embryos do not develop any lens structure, despite the fact that Rx is not expressed in the lens. This demonstrates that during normal mammalian development, retina-specific gene expression is necessary for lens formation. In this paper we show that lens formation can be restored in Rx-deficient embryos experimentally, by the elimination of beta-catenin expression in the head surface ectoderm. This suggests that beta-catenin is involved in lens specification either through Wnt signaling or through its function in cell adhesion. In contrast to lens formation, we demonstrate that the development of auxiliary eye structures does not depend on retina-specific gene expression or retinal morphogenesis. These results point to the existence of two separate developmental processes involved in the formation of the eye and its associated structures. One involved in the formation of the eyeball and the second involved in the formation of the auxiliary eye structures.


Assuntos
Proteínas do Olho/fisiologia , Olho/embriologia , Proteínas de Homeodomínio/fisiologia , Retina/embriologia , beta Catenina/fisiologia , Animais , Túnica Conjuntiva/citologia , Túnica Conjuntiva/embriologia , Olho/citologia , Proteínas do Olho/genética , Pálpebras/citologia , Pálpebras/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Homeodomínio/genética , Hibridização In Situ , Aparelho Lacrimal/citologia , Aparelho Lacrimal/embriologia , Cristalino/citologia , Cristalino/embriologia , Camundongos , Camundongos Knockout , Retina/citologia , beta Catenina/genética
20.
Genesis ; 46(3): 177-83, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18327772

RESUMO

In this article, we investigate the expression, regulation, and function of the zebrafish forkhead gene foxe3. In wild type embryos, foxe3 is first expressed in a crescent-shaped area at the anterior end of the prechordal plate, corresponding to the polster. At later stages, the hatching gland, the lens, and the anterior pituitary express this gene. Using morpholinos against the zinc finger Kruppel-like factor 4 (KLF4) we show that foxe3 is regulated differently in the polster and in the lens. In the absence of KLF4, expression of foxe3 in the polster is not activated, whereas in the lens placode the expression of KLF4 is not required for the transcription of foxe3. The expression of foxe3 is also regulated by the hedgehog and nodal signaling pathways. foxe3 expression is altered in the hedgehog pathway mutants iguana and you-too and the nodal pathway mutant cyclops. foxe3 function is necessary for the execution of lens-specific gene expression and lens morphogenesis, as the knockdown of foxe3 results in a loss of platelet-derived growth factor receptor alpha (pdgfralpha) expression and in the vacuolization of the lens.


Assuntos
Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Embrião não Mamífero , Proteínas do Olho/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cristalino/embriologia , Cristalino/metabolismo , Morfogênese/genética , Proteínas Mutantes/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteína Gli2 com Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...