Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Dev Neurobiol ; 75(9): 947-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25556712

RESUMEN

The histone modification H3K27me3 regulates transcription negatively, and Jmjd3 and Ezh2 demethylate and methylate H3K27me3 and H3K27, respectively. We demonstrated previously that Jmjd3 plays pivotal roles in the differentiation of subsets of bipolar (BP) cells by regulating H3K27me3 levels at the Bhlhb4 and Vsx1 loci, both of which are transcription factors essential for the maturation of BP cell subsets. In this study, we examined the role of Ezh2 in retinal development using retina-specific Ezh2 conditional knockout mice (Ezh2-CKO). The eyes of the Ezh2-CKO mice were microphthalemic, and the proliferation of retinal cells was diminished postnatally in Ezh2-CKO. Differentiation of all examined retinal subsets was observed with higher proportion of BP cell subsets, which was determined by immunostaining using specific retinal markers. The onsets of Müller glia and rod photoreceptor differentiation were accelerated. The expression of Bhlhb4 was increased in postnatal retinas, which was accompanied by the loss of H3K27me3 modifications at these genetic loci. Decreased expression of proneural genes in postnatal stage was observed. As reported previously in other Ezh2-KO tissues, increased expression of Arf/Ink4a was observed in the Ezh2-CKO retinas. The ectopic expression of Arf or Ink4a in the retina suppressed proliferation and increased apoptosis. In addition, earlier onset of Müller glia differentiation was observed in Ink4a-expressing cells. These results support an important role for histone H3K27me3 modification in regulating the proliferation and maturation of certain subsets of interneurons in the retina.


Asunto(s)
Proliferación Celular/fisiología , Neurogénesis/fisiología , Complejo Represivo Polycomb 2/metabolismo , Retina/crecimiento & desarrollo , Retina/fisiología , Animales , Animales Recién Nacidos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Interneuronas/patología , Interneuronas/fisiología , Ratones Noqueados , Neuroglía/patología , Neuroglía/fisiología , Retina/patología
2.
Biochimie ; 106: 68-74, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25127661

RESUMEN

The vertebrate lens undergoes organelle and nuclear degradation during lens development, allowing the lens to become transparent. DNase2b is an enzyme responsible for nuclear degradation in the mouse lens; however, dnase2b expression in zebrafish showed a distribution pattern that differed from that in mice. No zebrafish dnase2b was detected by reverse-transcription polymerase chain reaction until around 120 h postfertilization (hpf), suggesting that dnase2b is not expressed in the critical period for lens nuclear degradation, which corresponds to 56-74 hpf. However, public database searches have indicated that dnase1l3l is strongly and specifically expressed in embryonic zebrafish lens. Whole mount in situ hybridization showed that dnase1l3l expression began around 36 hpf and was found exclusively in the lens until the adult stage. Morpholino (MO)-dependent downregulation of dnase1l3l expression during early development in zebrafish led to the failure of nuclear degradation in the lens. Immunostaining of lens sections showed that expression of Pax6, Prox1 and ß-catenin was comparable to the control in the early stage of development in dnase1l3l-MO injected embryos. However, downregulation of expression of these genes in lens was not observed in dnase1l3l-MO-treated zebrafish at 72 hpf, suggesting that the lens development was halted. Taken together, we showed that dnase1l3l plays major roles in nuclear degradation in zebrafish lens development. No homologous gene was found in other species in public databases, suggesting that dnase1l3l developed and acquired its function specifically in zebrafish.


Asunto(s)
Desoxirribonucleasas/genética , Regulación del Desarrollo de la Expresión Génica , Cristalino/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Desoxirribonucleasas/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Cristalino/citología , Cristalino/embriología , Masculino , Ratones Endogámicos ICR , Morfolinos/genética , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(10): 3751-6, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24572572

RESUMEN

Di- and trimethylation of lysine 27 on histone H3 (H3K27me2/3) is an important gene repression mechanism. H3K27me2/3-specific demethylase, Jmjd3, was expressed in the inner nuclear layer during late retinal development. In contrast, H3K27 methyltransferase, Ezh2, was highly expressed in the embryonic retina but its expression decreased rapidly after birth. Jmjd3 loss of function in the developing retina resulted in failed differentiation of PKC-positive bipolar cell subsets (rod-ON-BP) and reduced transcription factor Bhlhb4 expression, which is critical for the differentiation of rod-ON-BP cells. Overexpression of Bhlhb4, but not of other BP cell-related genes, such as transcription factors Neurod and Chx10, in Jmjd3-knockdown retina rescued loss of PKC-positive BP cells. Populations of other retinal cell subsets were not significantly affected. In addition, proliferation activity and apoptotic cell number during retinal development were not affected by the loss of Jmjd3. Levels of histone H3 trimethyl Lys27 (H3K27me3) in the Bhlhb4 locus were lower in Islet-1-positive BP cells and amacrine cells than in the Islet-1-negative cell fraction. The Islet-1-negative cell fraction consisted mainly of photoreceptors, suggestive of lineage-specific demethylation of H3K27me3 in the Bhlhb4 locus. We propose that lineage-specific H3K27me3 demethylation of critical gene loci by spatiotemporal-specific Jmjd3 expression is required for appropriate maturation of retinal cells.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Bipolares de la Retina/fisiología , Anticuerpos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Electroporación , Citometría de Flujo , Humanos , Hibridación in Situ , Metilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Bipolares de la Retina/citología
4.
Dev Neurobiol ; 74(4): 426-37, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24124169

RESUMEN

Retinal progenitor cells alter their properties over the course of development, and sequentially produce different sub-populations of retinal cells. We had previously found that early and late retinal progenitor cell populations can be distinguished by their surface antigens, SSEA-1 and c-kit, respectively. Using DNA microarray analysis, we examined the transcriptomes of SSEA-1 positive cells at E14, and c-kit positive, and c-kit negative cells at P1. By comparing data, we identified genes specifically expressed in c-kit positive late retinal progenitor cells. The previous literature suggests that most of the c-kit positive cell-specific genes are related to glia differentiation in brain or are expressed in Müller glia. Since Notch signaling promotes Müller glia differentiation in retina, we examined the effects of gain- and loss-of-Notch signaling on expression of these genes and found that all the genes were positively affected by Notch signaling. Finally, we screened the genes for their function in retinal development by shRNA-based suppression in retinal explants. In about half the genes, Müller glia differentiation was perturbed when their expression was suppressed. Taken together, these results show that at P1, c-kit positive retinal progenitor cells, which include Müller glia precursor cells, are enriched for genes related to glial differentiation. We propose analysis of purified subsets of retinal cells as a powerful tool to elucidate the molecular basis of retinal development.


Asunto(s)
Diferenciación Celular , Células Ependimogliales/fisiología , Retina/crecimiento & desarrollo , Retina/fisiología , Animales , Animales Recién Nacidos , Electroporación , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Técnicas In Vitro , Antígeno Lewis X/metabolismo , Ratones , Ratones Endogámicos ICR , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Interferente Pequeño , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/fisiología , Transcriptoma
5.
PLoS One ; 8(8): e72065, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24058409

RESUMEN

Cone photopigments (opsins) are crucial elements of, and the first detection module in, color vision. Individual opsins have different wavelength sensitivity patterns, and the temporal and spatial expression patterns of opsins are unique and stringently regulated. Long and middle wavelength-sensitive (L/M) opsins are of the same phylogenetic type. Although the roles of thyroid hormone/TRß2 and COUP-TFs in the transcriptional regulation of L/M opsins have been explored, the detailed mechanisms, including the target sequence in the enhancer of L/M opsins, have not been revealed. We aimed to reveal molecular mechanisms of L/M opsins in vertebrates. Using several human red opsin enhancer/promoter-luciferase reporter constructs, we found that TRß2 increased luciferase activities through the 5'-UTR and intron 3-4 region, whereas the presence of T3 affected only the intron 3-4 region-dependent luciferase activity. Furthermore, COUP-TFII suppressed intron 3-4 region-dependent luciferase activities. However, luciferase expression driven by the mouse M opsin intron 3-4 region was only slightly increased by TRß2, and rather enhanced by COUP-TFII. To determine whether these differential responses reflect differences between primates and rodents, we examined the enhancer/promoter region of the red opsin of the common marmoset. Interestingly, while TRß2 increased 5'-UTR- or intron 3-4 region-driven luciferase expression, as observed for the human red opsin, expression of the latter luciferase was not suppressed by COUP-TFII. In fact, immunostaining of common marmoset retinal sections revealed expression of COUP-TFII and red opsin in the cone cells.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Opsinas de Bastones/genética , Receptores beta de Hormona Tiroidea/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/metabolismo , Ratones
6.
Development ; 140(4): 740-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23318640

RESUMEN

Sry-related HMG box (Sox) proteins, Sox11 and Sox4 are members of the SoxC subtype. We found that Sox11 was strongly expressed in early retinal progenitor cells and that Sox4 expression began around birth, when expression of Sox11 subsided. To analyze the roles of Sox11 and Sox4 in retinal development, we perturbed their expression patterns in retinal explant cultures. Overexpression of Sox11 and Sox4 in retinal progenitors resulted in similar phenotypes: an increased number of cone cells and dramatically decreased numbers of rod cells and Müller glia. Birth-date analysis showed that cone cells were produced at a later developmental stage than that in which cone genesis normally occurs. Sox11-knockout retinas showed delayed onset and progress of differentiation of subsets of retinal cells during the embryonic period. After birth, retinal differentiation took place relatively normally, probably because of the redundant activity of Sox4, which starts to be expressed around birth. Overexpression and loss-of-function analysis failed to provide any evidence that Sox11 and Sox4 directly regulate the transcription of genes crucial to the differentiation of subsets of retinal cells. However, histone H3 acetylation of some early proneural genes was reduced in knockout retina. Thus, Sox11 may create an epigenetic state that helps to establish the competency to differentiate. Taking our findings together, we propose that the sequential expression of Sox11 and Sox4 during retinogenesis leads to the fine adjustment of retinal differentiation by helping to establish the competency of retinal progenitors.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Fotorreceptoras de Vertebrados/fisiología , Retina/citología , Retina/embriología , Factores de Transcripción SOXC/metabolismo , Células Madre/fisiología , Acetilación , Animales , Bromodesoxiuridina , Inmunoprecipitación de Cromatina , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Histonas/metabolismo , Hibridación in Situ , Ratones , Células 3T3 NIH , Oligonucleótidos/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factores de Tiempo
7.
FEBS Lett ; 587(4): 358-63, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23313252

RESUMEN

Sox11 and Sox4 play critical roles in retinal development, during which they display specific and unique expression patterns. The expression of Sox11 and Sox4 is temporally sequential, albeit spatially overlapping in some retinal subtypes. Gain-of-function and loss-of-function analyses suggested that Notch signaling suppresses Sox4 expression in the early developing retina but not during the later period of development. The levels of histone H3-acetylation and H3-lysine 4 tri-methylation at the Sox11 locus declined during development, as did the levels of Sox11. A similar but less marked change was seen for Sox4. For both genes, histone H3-lysine 27 methylation was low during development and increased markedly in the adult.


Asunto(s)
Envejecimiento , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Retina/metabolismo , Factores de Transcripción SOXC/metabolismo , Acetilación , Animales , Células Cultivadas , Metilación de ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Metilación , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Retina/citología , Retina/crecimiento & desarrollo , Factores de Transcripción SOXC/genética , Transducción de Señal , Técnicas de Cultivo de Tejidos
8.
Appl Biochem Biotechnol ; 166(1): 64-71, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22052544

RESUMEN

MicroRNA-140 (miR-140) is specifically expressed in developing cartilage tissues. We have previously reported that miR-140 plays an important role during palatal cartilage development by modulating platelet-derived growth factor receptor alpha (pdgfra) in zebrafish. However, the regulatory mechanism of miR-140 in cartilage is still unknown. Using developing zebrafish, sox9a mutant (sox9a-/-) and sox9b mutant (sox9b-/-) zebrafish and SOX9 small interfering RNA in human chondrocytes, T/C-28 cells, we found that miR-140 is regulated by the cartilage master transcription regulator Sox9 in zebrafish and mammalian cells.


Asunto(s)
Cartílago/crecimiento & desarrollo , MicroARNs/genética , MicroARNs/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Sitios de Unión , Cartílago/metabolismo , Condrocitos/metabolismo , Condrogénesis/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Filogenia , Unión Proteica , ARN Interferente Pequeño/genética , Ubiquitina-Proteína Ligasas/genética , Pez Cebra/genética
9.
Biochimie ; 93(6): 1037-46, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21396426

RESUMEN

The region-specific homeotic gene spalt (sal) gene plays a critical role in Drosophila development. The mammalian Sal homologous genes contain four members, and Sall3 is mainly expressed in horizontal cells. In the developing retinas of Sall3 knockout (KO) mice until around birth, horizontal precursor cells developed with comparable numbers and position; the horizontal cell marker NF160 was expressed weakly and neurite-like structure had once formed. Since Sall3-KO mice die at postnatal day 1, subsequent retinal development was examined by in vitro retinal explant culture. In the Sall3-KO retina culture, the expression of NF160 was abrogated, and neurite extension was not observed. Furthermore, Sall3-KO horizontal precursors were initially localized at the appropriate horizontal positions, but eventually moved to an abnormal site in the outer nuclear layer. Overexpression of Sall3 in retinal progenitors did not induce differentiation of retinal progenitor cells into the horizontal cell-fate, but enhanced NF160 expression and neurite extension. In addition, differentiation into Müller glia was promoted, and rod cells were severely suppressed without perturbing proliferation. In conclusion, Sall3 may not be involved in horizontal cell-fate determination, but rather functions to instruct terminal differentiation of horizontal cells and to maintain NF160 expression.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Proteínas de Neurofilamentos/metabolismo , Células Horizontales de la Retina/metabolismo , Factores de Transcripción/metabolismo , Animales , Calbindinas , Caspasa 3/metabolismo , Muerte Celular/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Conexinas/genética , Conexinas/metabolismo , Proteínas del Ojo/metabolismo , Eliminación de Gen , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Neuritas/metabolismo , Proteínas de Neurofilamentos/genética , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Retina/citología , Retina/embriología , Retina/metabolismo , Células Horizontales de la Retina/citología , Proteína G de Unión al Calcio S100/metabolismo , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética , Transcripción Genética
10.
Invest Ophthalmol Vis Sci ; 52(6): 3008-17, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21273552

RESUMEN

PURPOSE: Much attention has been paid to the roles of microRNA in developmental and biological processes. Dicer plays essential roles in cell survival and proliferation in various organs. We examined the role of Dicer in retinal development using retina-specific conditional knockout of Dicer in mice. METHODS: Dkk3-Cre expressed the Cre gene in retinal progenitor cells from an early embryonic stage. The authors analyzed Dkk-Cre/Dicer-flox (Dicer-CKO) mice for their survival, proliferation, and differentiation. To analyze the role of Dicer in later stages of retinal development, a Cre expression plasmid was introduced into the neonatal retina by electroporation, and retinal differentiation was examined. RESULTS: Dicer-CKO mice were born at the numbers we expected, based on Mendelian genetics, but their eyes never opened. Massive death of retinal progenitor cells occurred during embryogenesis, resulting in microphthalmia, and most retinal cells had disappeared by postnatal day 14. In vitro reaggregation culture of Dicer-CKO retinal cells showed that cell death and the suppression of proliferation by Dicer inactivation occurred in a cell-autonomous manner. Cell differentiation markers were expressed in the Dicer-CKO retina; however, these cells localized abnormally, and the inner plexiform layer was absent, suggesting that cell migration and morphologic differentiation, especially process extension, were perturbed. Forced neonatal expression of Cre induced apoptosis and affected the expression of differentiation markers. CONCLUSIONS: Taken together, these results show that Dicer is essential during early retinal development.


Asunto(s)
Diferenciación Celular , ARN Helicasas DEAD-box/fisiología , Microftalmía/patología , Retina/embriología , Degeneración Retiniana/patología , Ribonucleasa III/fisiología , Células Madre/patología , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Electroporación , Desarrollo Embrionario , Eliminación de Gen , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Microftalmía/metabolismo , Plásmidos , Retina/patología , Degeneración Retiniana/metabolismo
11.
Exp Eye Res ; 90(1): 49-56, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19766631

RESUMEN

Chicken ovalbumin upstream promoter transcription factors (COUP-TFs) are members of the steroid/thyroid hormone receptor superfamily. We have shown that two homologous COUP-TF genes, COUP-TFI and COUP-TFII, are expressed in developing mouse retina with a unique gradient along the dorsal-ventral axis. In this work, we aimed to characterize the detailed expression patterns of COUP-TFs in mature retina. Their functions in retinal progenitor cell differentiation into subtypes of mature retinal cells were also examined. Immunostaining of frozen mouse retinal sections with antibodies against COUP-TFs and markers for retinal subtypes revealed that COUP-TFI and -TFII are expressed in amacrine cells, especially in a glycinergic subtype in mature mouse retina. Forced expression of COUP-TFI and -TFII in mouse retinal explant culture by retrovirus-mediated gene transfer promoted amacrine and cone photoreceptor cell differentiation, whereas that of rod photoreceptors decreased. Cell proliferation and apoptosis were not affected by the perturbation of COUP-TFI and -TFII expression levels. Using the Y79 retinoblastoma cell line, we observed that COUP-TFI and -TFII suppressed the transcriptional activation of the Nrl gene. We then analyzed one another member of COUP-TF transcription factors, COUP-TFgamma, whose structure is relatively distant from those of COUP-TFI and -TFII. It is expressed mainly in horizontal cells and has weak activity in inducing amacrine cells when COUP-TFgamma was ectopically expressed in retinal explants. In summary, we found that COUP-TFI and -TFII play roles in amacrine cell differentiation, and COUP-TFgamma has distinct expression pattern and roles during retinal development.


Asunto(s)
Células Amacrinas/metabolismo , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP I/genética , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Retina/citología , Células Madre/citología , Animales , Apoptosis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proliferación Celular , Proteínas del Ojo/genética , Citometría de Flujo , Ratones , Ratones Endogámicos ICR , Plásmidos , Neoplasias de la Retina/genética , Retinoblastoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas
12.
J Neurosci ; 29(40): 12401-11, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19812316

RESUMEN

Cone photopigments, known as opsins, are pivotal elements and the first detection module used in color vision. In mice, cone photoreceptors are distributed throughout the retina, and short-wavelength (S) and medium-wavelength (M) opsins have unique expression patterns in the retina with a gradient along the dorsoventral axis; however, the mechanisms regulating the spatial patterning of cone opsin expression have not been well documented. The purpose of this study was to define the mechanisms regulating the spatial patterning of cone opsin expression. By analyzing knock-outs for bone morphogenetic protein (BMP) signaling, we found an essential role for BMP in forming cone opsin expression patterns in the retina; however, BMP signaling is activated only transiently in the dorsal half of the retina during early retinal development. Thus, BMP is not likely to play a direct role in opsin gene expression, which starts at a later stage of retinal development. We identified the chicken ovalbumin upstream promoter-transcription factor (COUP-TF) nuclear receptor as a link between BMP and opsin expression. BMP signaling is essential for the correct dorsoventral spatial expression of COUP-TFI and COUP-TFII. Through gain- and loss-of-function analyses, we found that both COUP-TFI and COUP-TFII are required to suppress S-opsin expression in the dorsal retina but that only COUP-TFI plays an essential role in suppressing M-opsin expression in the ventral retina. Based on these findings, we propose a new molecular cascade involving BMP and COUP-TFs that conveys dorsoventral information to direct the expression of cone opsins during retinal development.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factores de Transcripción COUP/metabolismo , Opsinas de los Conos/metabolismo , Retina/embriología , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Tipificación del Cuerpo/fisiología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratas , Ratas Wistar , Transducción de Señal
13.
Exp Eye Res ; 89(4): 549-58, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19490914

RESUMEN

Although Müller glial cells play pivotal roles in the vertebrate retina, the regulation of their development is poorly understood. While Notch-Hes5 signaling has been shown to be involved in this developmental process, the presence of Müller glial cells in Hes5-deficient mice suggests the involvement of other molecules. We found that two group E Sox genes, Sox8 and Sox9, are expressed in proliferating progenitors and then exclusively in Müller glial cells in mouse retina. Knocking-down Sox8 and Sox9 by shRNA significantly reduced the population of Müller glial cells and relatively increased the proportion of rod photoreceptors, suggesting that the Sox genes play roles in the specification of Müller glial cells. Using an activated form of Notch and the gamma-secretase inhibitor DAPT, we also found that Notch signaling regulates the transcription of Sox8 and Sox9. This is the first evidence that group E Sox genes play important roles in the developing vertebrate retina.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Neuroglía/metabolismo , Receptores Notch/metabolismo , Retina/embriología , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXE/genética , Transducción de Señal , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Western Blotting , Técnicas de Cultivo de Célula , Dipéptidos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Técnicas para Inmunoenzimas , Hibridación in Situ , Ratones , Ratones Endogámicos ICR , Plásmidos , ARN Mensajero/metabolismo , Retina/metabolismo , Transfección
14.
J Exp Zool B Mol Dev Evol ; 312(8): 827-40, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19488987

RESUMEN

Forkhead transcription factor (Fox) e1 is a causative gene for Bamforth-Lazarus syndrome, which is characterized by hypothyroidism and cleft palate. Applying degenerate polymerase chain reaction using primers specific for the conserved forkhead domain, we identified zebrafish foxe1 (foxe1). Foxe1 is expressed in the thyroid, pharynx, and pharyngeal skeleton during development; strongly expressed in the gill and weakly expressed in the brain, eye, and heart in adult zebrafish. A loss of function of foxe1 by morpholino antisense oligo (MO) exhibited abnormal craniofacial development, shortening of Meckel's cartilage and the ceratohyals, and suppressed chondrycytic proliferation. However, at 27 hr post fertilization, the foxe1 MO-injected embryos showed normal dlx2, hoxa2, and hoxb2 expression, suggesting that the initial steps of pharyngeal skeletal development, including neural crest migration and specification of the pharyngeal arch occurred normally. In contrast, at 2 dpf, a severe reduction in the expression of sox9a, colIIaI, and runx2b, which play roles in chondrocytic proliferation and differentiation, was observed. Interestingly, fgfr2 was strongly upregulated in the branchial arches of the foxe1 MO-injected embryos. Unlike Foxe1-null mice, normal thyroid development in terms of morphology and thyroid-specific marker expression was observed in foxe1 MO-injected zebrafish embryos. Taken together, our results indicate that Foxe1 plays an important role in chondrogenesis during development of the pharyngeal skeleton in zebrafish, probably through regulation of fgfr2 expression. Furthermore, the roles reported for FOXE1 in mammalian thyroid development may have been acquired during evolution.


Asunto(s)
Cartílago/embriología , Factores de Transcripción/fisiología , Pez Cebra/embriología , Animales , Secuencia de Bases , Cartilla de ADN , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándula Tiroides/metabolismo , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética
15.
Stem Cells ; 26(12): 3162-71, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18787210

RESUMEN

Ciliary epithelium (CE), which consists of nonpigmented and pigmented layers, develops from the optic vesicle. However, the molecular mechanisms underlying CE development have not been closely examined, in part because cell-surface markers suitable for specific labeling of subregions of the retina were unknown. Here, we identified CD138/syndecan-1 and stage specific embryonic antigen-1 (SSEA-1) CD15 as cell-surface antigens marking nonpigmented and pigmented CE, respectively. During retinal development, both CD138 and SSEA-1 were expressed in the early stage, and segregation of these markers in the tissue began at around embryonic day (E) 10. As a result, CD138-positive (CD138+) cells were found at the most distal tip of the retina, and SSEA-1+ cells were found in the periphery adjacent to the area of CD138 expression. In vitro characterization of isolated CD138+ or SSEA-1+ cell subpopulations revealed that CD138+ cells lose their retinal progenitor characteristics between E13 and E16, suggesting that they commit to becoming nonpigmented CE cells within this period. By in vivo mouse models, we found that stabilized beta-catenin expanded the area of CD138+ nonpigmented CE and that elimination of beta-catenin inhibited development of nonpigmented CE cells. These findings are the first to use cell-surface markers to ascertain the spatial and temporal transitions that occur in developing CE.


Asunto(s)
Cuerpo Ciliar/embriología , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica , Antígeno Lewis X/biosíntesis , Sindecano-1/biosíntesis , Proteínas Wnt/metabolismo , Alelos , Animales , Diferenciación Celular , Membrana Celular/metabolismo , Cuerpo Ciliar/metabolismo , Citometría de Flujo , Antígeno Lewis X/fisiología , Ratones , Ratones Endogámicos C57BL , Retina/citología , Retina/embriología , Sindecano-1/fisiología
16.
Mol Vis ; 14: 1623-30, 2008 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-18776950

RESUMEN

PURPOSE: Glycoprotein m6a (M6a) is a cell-surface glycoprotein that belongs to the myelin proteolipid protein family. M6a is expressed mainly in the nervous system, and its expression and function in mammalian retina have not been described. Using proteomics analysis of mouse retinal membrane fractions, we identified M6a as a retinal membrane protein that is strongly expressed at embryonic stages. Our aim was to reveal the function of M6a in development of mouse retina in this work. METHODS: Detailed expression pattern of M6a was examined by immunostaining using frozen sections of mouse retina obtained at various developmental stages. For functional analysis of M6a in mouse retinal development, we performed retorovirus-mediated overexpression of M6a in mouse retinal explant culture. Then, cell differentiation, proliferation and structural maturation of the cells were examined. RESULTS: M6a transcripts were strongly expressed in embryonic retina. After completion of retinal differentiation, the level of expression decreased as mouse development progressed. Immunohistochemistry showed that in the immature mouse retina, M6a was strongly expressed in the axons of retinal ganglion cells. After birth, M6a expression was confined to the inner plexiform layer, and finally, to the inner and outer plexiform layers of adult mouse retina. M6a expression was completely paralleled by that of the synaptic marker, synaptophysin. Mouse retinal progenitor cells that overexpressed M6a following retrovirus-mediated gene transfer were subjected to in vitro explant or monolayer cultures. The neurite outgrowth of M6a-overexpressing retinal cells was strikingly enhanced, although M6a did not affect differentiation and proliferation. CONCLUSIONS: These results suggest that M6a plays a role in retinal development by regulating neurites, and it may also function to modulate synaptic activities in the adult retina.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Retina/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos ICR , Mitosis , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas , Retina/citología , Retina/embriología , Células Madre/citología , Células Madre/metabolismo , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA