Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Development ; 142(23): 4092-106, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26628093

RESUMEN

Retinal development requires precise temporal and spatial coordination of cell cycle exit, cell fate specification, cell migration and differentiation. When this process is disrupted, retinoblastoma, a developmental tumor of the retina, can form. Epigenetic modulators are central to precisely coordinating developmental events, and many epigenetic processes have been implicated in cancer. Studying epigenetic mechanisms in development is challenging because they often regulate multiple cellular processes; therefore, elucidating the primary molecular mechanisms involved can be difficult. Here we explore the role of Brg1 (Smarca4) in retinal development and retinoblastoma in mice using molecular and cellular approaches. Brg1 was found to regulate retinal size by controlling cell cycle length, cell cycle exit and cell survival during development. Brg1 was not required for cell fate specification but was required for photoreceptor differentiation and cell adhesion/polarity programs that contribute to proper retinal lamination during development. The combination of defective cell differentiation and lamination led to retinal degeneration in Brg1-deficient retinae. Despite the hypocellularity, premature cell cycle exit, increased cell death and extended cell cycle length, retinal progenitor cells persisted in Brg1-deficient retinae, making them more susceptible to retinoblastoma. ChIP-Seq analysis suggests that Brg1 might regulate gene expression through multiple mechanisms.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Retina/metabolismo , Retinoblastoma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Apoptosis , Tipificación del Cuerpo , Adhesión Celular , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Ratones , Microftalmía/genética , Retina/patología , Factores de Tiempo , Transgenes
2.
BMC Cancer ; 17(1): 434, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28633655

RESUMEN

BACKGROUND: Retinoblastoma (Rb) is the most common primary intraocular tumor in children. Local treatment of the intraocular disease is usually effective if diagnosed early; however advanced Rb can metastasize through routes that involve invasion of the choroid, sclera and optic nerve or more broadly via the ocular vasculature. Metastatic Rb patients have very high mortality rates. While current therapy for Rb is directed toward blocking tumor cell division and tumor growth, there are no specific treatments targeted to block Rb metastasis. Two such targets are matrix metalloproteinases-2 and -9 (MMP-2, -9), which degrade extracellular matrix as a prerequisite for cellular invasion and have been shown to be involved in other types of cancer metastasis. Cancer Clinical Trials with an anti-MMP-9 therapeutic antibody were recently initiated, prompting us to investigate the role of MMP-2, -9 in Rb metastasis. METHODS: We compare MMP-2, -9 activity in two well-studied Rb cell lines: Y79, which exhibits high metastatic potential and Weri-1, which has low metastatic potential. The effects of inhibitors of MMP-2 (ARP100) and MMP-9 (AG-L-66085) on migration, angiogenesis, and production of immunomodulatory cytokines were determined in both cell lines using qPCR, and ELISA. Cellular migration and potential for invasion were evaluated by the classic wound-healing assay and a Boyden Chamber assay. RESULTS: Our results showed that both inhibitors had differential effects on the two cell lines, significantly reducing migration in the metastatic Y79 cell line and greatly affecting the viability of Weri-1 cells. The MMP-9 inhibitor (MMP9I) AG-L-66085, diminished the Y79 angiogenic response. In Weri-1 cells, VEGF was significantly reduced and cell viability was decreased by both MMP-2 and MMP-9 inhibitors. Furthermore, inhibition of MMP-2 significantly reduced secretion of TGF-ß1 in both Rb models. CONCLUSIONS: Collectively, our data indicates MMP-2 and MMP-9 drive metastatic pathways, including migration, viability and secretion of angiogenic factors in Rb cells. These two subtypes of matrix metalloproteinases represent new potential candidates for targeted anti-metastatic therapy for Rb.


Asunto(s)
Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Neovascularización Patológica/tratamiento farmacológico , Retinoblastoma/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Retinoblastoma/genética , Retinoblastoma/patología , Factor A de Crecimiento Endotelial Vascular/genética
3.
Proc Natl Acad Sci U S A ; 108(52): 21111-6, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160703

RESUMEN

Neuronal differentiation with respect to the acquisition of synaptic competence needs to be regulated precisely during neurogenesis to ensure proper formation of circuits at the right place and time in development. This regulation is particularly important for synaptic triads among photoreceptors, horizontal cells (HCs), and bipolar cells in the retina, because HCs are among the first cell types produced during development, and bipolar cells are among the last. HCs undergo a dramatic transition from vertically oriented neurites that form columnar arbors to overlapping laminar dendritic arbors with differentiation. However, how this process is regulated and coordinated with differentiation of photoreceptors and bipolar cells remains unknown. Previous studies have suggested that the retinoblastoma (Rb) tumor suppressor gene may play a role in horizontal cell differentiation and synaptogenesis. By combining genetic mosaic analysis of individual synaptic triads with neuroanatomic analyses and multiphoton live imaging of developing HCs, we found that Rb plays a cell-autonomous role in the reorganization of horizontal cell neurites as they differentiate. Aberrant vertical processes in Rb-deficient HCs form ectopic synapses with rods in the outer nuclear layer but lack bipolar dendrites. Although previous reports indicate that photoreceptor abnormalities can trigger formation of ectopic synapses, our studies now demonstrate that defects in a postsynaptic partner contribute to the formation of ectopic photoreceptor synapses in the mammalian retina.


Asunto(s)
Diferenciación Celular/fisiología , Dendritas/fisiología , Neurogénesis/fisiología , Células Horizontales de la Retina/citología , Proteína de Retinoblastoma/metabolismo , Sinapsis/fisiología , Animales , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Proteína de Retinoblastoma/genética
4.
Blood ; 116(8): 1377-85, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20421451

RESUMEN

Vascular endothelial growth factor (VEGF) appears to be an important mediator of pathologic retinal angiogenesis. In understanding the mechanisms of pathologic retinal neovascularization, we found that VEGF activates PLD1 in human retinal microvascular endothelial cells, and this event is dependent on Src. In addition, VEGF activates protein kinase C-gamma (PKCgamma) via Src-dependent PLD1 stimulation. Inhibition of Src, PLD1, or PKCgamma via pharmacologic, dominant negative mutant, or siRNA approaches significantly attenuated VEGF-induced human retinal microvascular endothelial cell migration, proliferation, and tube formation. Hypoxia also induced Src-PLD1-PKCgamma signaling in retina, leading to retinal neovascularization. Furthermore, siRNA-mediated down-regulation of VEGF inhibited hypoxia-induced Src-PLD1-PKCgamma activation and neovascularization. Blockade of Src-PLD1-PKCgamma signaling via the siRNA approach also suppressed hypoxia-induced retinal neovascularization. Thus, these observations demonstrate, for the first time, that Src-dependent PLD1-PKCgamma activation plays an important role in pathologic retinal angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Fosfolipasa D/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Retina/metabolismo , Neovascularización Retiniana , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Fosforilación , Retina/patología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Nature ; 444(7115): 61-6, 2006 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-17080083

RESUMEN

Most human tumours have genetic mutations in their Rb and p53 pathways, but retinoblastoma is thought to be an exception. Studies suggest that retinoblastomas, which initiate with mutations in the gene retinoblastoma 1 (RB1), bypass the p53 pathway because they arise from intrinsically death-resistant cells during retinal development. In contrast to this prevailing theory, here we show that the tumour surveillance pathway mediated by Arf, MDM2, MDMX and p53 is activated after loss of RB1 during retinogenesis. RB1-deficient retinoblasts undergo p53-mediated apoptosis and exit the cell cycle. Subsequently, amplification of the MDMX gene and increased expression of MDMX protein are strongly selected for during tumour progression as a mechanism to suppress the p53 response in RB1-deficient retinal cells. Our data provide evidence that the p53 pathway is inactivated in retinoblastoma and that this cancer does not originate from intrinsically death-resistant cells as previously thought. In addition, they support the idea that MDMX is a specific chemotherapeutic target for treating retinoblastoma.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular , Muerte Celular , División Celular , Daño del ADN , Amplificación de Genes/genética , Humanos , Imidazoles/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Piperazinas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Retina/metabolismo , Retinoblastoma/genética , Retinoblastoma/patología , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(16): 6685-90, 2009 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-19346468

RESUMEN

It was previously reported that the ciliary epithelium (CE) of the mammalian eye contains a rare population of cells that could produce clonogenic self-renewing pigmented spheres in culture. Based on their ability to up-regulate genes found in retinal neurons, it was concluded that these sphere-forming cells were retinal stem cells. This conclusion raised the possibility that CE-derived retinal stem cells could help to restore vision in the millions of people worldwide who suffer from blindness associated with retinal degeneration. We report here that human and mouse CE-derived spheres are made up of proliferating pigmented ciliary epithelial cells rather than retinal stem cells. All of the cells in the CE-derived spheres, including the proliferating cells, had molecular, cellular, and morphological features of differentiated pigmented CE cells. These differentiated cells ectopically expressed nestin when exposed to growth factors and low levels of pan-neuronal markers such as beta-III-tubulin. Although the cells aberrantly expressed neuronal markers, they retained their pigmented CE cell morphology and failed to differentiate into retinal neurons in vitro or in vivo. Our results provide an example of a differentiated cell type that can form clonogenic spheres in culture, self-renew, express progenitor cell markers, and initiate neuronal differentiation that is not a stem or progenitor cell. More importantly, our findings highlight the importance of shifting the focus away from studies on CE-derived spheres for cell-based therapies to restore vision in the degenerating retina and improving techniques for using ES cells or retinal precursor cells.


Asunto(s)
Cuerpo Ciliar/citología , Células Epiteliales/citología , Pigmentación , Retina/citología , Células Madre/citología , Adulto , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Cuerpo Ciliar/ultraestructura , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley
7.
Nat Commun ; 12(1): 4535, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315877

RESUMEN

Retinoblastoma is a childhood cancer of the developing retina that initiates with biallelic inactivation of the RB1 gene. Children with germline mutations in RB1 have a high likelihood of developing retinoblastoma and other malignancies later in life. Genetically engineered mouse models of retinoblastoma share some similarities with human retinoblastoma but there are differences in their cellular differentiation. To develop a laboratory model of human retinoblastoma formation, we make induced pluripotent stem cells (iPSCs) from 15 participants with germline RB1 mutations. Each of the stem cell lines is validated, characterized and then differentiated into retina using a 3-dimensional organoid culture system. After 45 days in culture, the retinal organoids are dissociated and injected into the vitreous of eyes of immunocompromised mice to support retinoblastoma tumor growth. Retinoblastomas formed from retinal organoids made from patient-derived iPSCs have molecular, cellular and genomic features indistinguishable from human retinoblastomas. This model of human cancer based on patient-derived iPSCs with germline cancer predisposing mutations provides valuable insights into the cellular origins of this debilitating childhood disease as well as the mechanism of tumorigenesis following RB1 gene inactivation.


Asunto(s)
Organoides/patología , Retina/patología , Retinoblastoma/patología , Células Madre/patología , Adulto , Diferenciación Celular , Línea Celular , Epigénesis Genética , Exones/genética , Femenino , Genoma Humano , Mutación de Línea Germinal/genética , Humanos , Imagenología Tridimensional , Células Madre Pluripotentes Inducidas/metabolismo , Retinoblastoma/genética , Proteína de Retinoblastoma/genética
8.
Cancer Res ; 67(6): 2701-11, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363591

RESUMEN

Retinoblastomas initiate in the developing retina in utero and are diagnosed during the first few years of life. We have recently generated a series of knockout mouse models of retinoblastoma that recapitulate the timing, location, and progression of human retinoblastoma. One of the most important benefits of these preclinical models is that we can study the earliest stages of tumor initiation and expansion. This is not possible in human retinoblastoma because tumors initiate in utero and are not diagnosed until they are at an advanced stage. We found that mouse retinoblastoma cells exhibit a surprising degree of differentiation, which has not been previously reported for any neural tumor. Early-stage mouse retinoblastoma cells express proteins found normally in retinal plexiform layers. They also extend neurites and form synapses. All of these features, which were characterized by immunostaining, Golgi-Cox staining, scanning electron microscopy, and transmission electron microscopy, suggest that mouse retinoblastoma cells resemble amacrine/horizontal cells from the retina. As late-stage retinoblastoma cells expand and invade the surrounding tissue, they lose their differentiated morphology and become indistinguishable from human retinoblastomas. Taken together, our data suggest that neuronal differentiation is a hallmark of early-stage retinoblastoma and is lost as cells become more aggressive and invasive. We also show that rosette formation is not a hallmark of retinoblastoma differentiation, as previously believed. Instead, rosette formation reflects extensive cell-cell contacts between retinoblastoma cells in both early-stage (differentiated) and late-stage (dedifferentiated) tumors.


Asunto(s)
Neuronas/patología , Retinoblastoma/patología , Sinapsis/patología , Animales , Cámara Anterior/patología , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica , Neuritas/patología , Neuronas/ultraestructura , Retina/patología , Retinoblastoma/genética , Retinoblastoma/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/ultraestructura , Cuerpo Vítreo/patología
9.
Neuron ; 104(3): 512-528.e11, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31493975

RESUMEN

More than 8,000 genes are turned on or off as progenitor cells produce the 7 classes of retinal cell types during development. Thousands of enhancers are also active in the developing retinae, many having features of cell- and developmental stage-specific activity. We studied dynamic changes in the 3D chromatin landscape important for precisely orchestrated changes in gene expression during retinal development by ultra-deep in situ Hi-C analysis on murine retinae. We identified developmental-stage-specific changes in chromatin compartments and enhancer-promoter interactions. We developed a machine learning-based algorithm to map euchromatin and heterochromatin domains genome-wide and overlaid it with chromatin compartments identified by Hi-C. Single-cell ATAC-seq and RNA-seq were integrated with our Hi-C and previous ChIP-seq data to identify cell- and developmental-stage-specific super-enhancers (SEs). We identified a bipolar neuron-specific core regulatory circuit SE upstream of Vsx2, whose deletion in mice led to the loss of bipolar neurons.


Asunto(s)
Eucromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Heterocromatina/metabolismo , Retina/embriología , Células Bipolares de la Retina/metabolismo , Animales , Cromatina/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Aprendizaje Automático , Ratones , Lámina Nuclear/metabolismo , Regiones Promotoras Genéticas , RNA-Seq , Receptores Citoplasmáticos y Nucleares/genética , Retina/citología , Retina/metabolismo , Retina/ultraestructura , Células Bipolares de la Retina/citología , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/genética , Receptor de Lamina B
10.
Cell Rep ; 22(10): 2601-2614, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514090

RESUMEN

Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.


Asunto(s)
Reprogramación Celular , Metilación de ADN , Histonas/metabolismo , Organogénesis , Organoides/crecimiento & desarrollo , Procesamiento Proteico-Postraduccional , Retina/citología , Retina/metabolismo , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Núcleo Celular/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Regiones Promotoras Genéticas/genética
11.
Invest Ophthalmol Vis Sci ; 48(11): 4930-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17962441

RESUMEN

PURPOSE: To examine for the expression of 15-lipoxygenase 1 (15-LOX1) and 15-LOX2 in human retinal microvascular endothelial cells (HRMVECs) and study the role of arachidonic acid metabolites of these enzymes in angiogenesis. METHODS: Quantitative RT-PCR and reverse-phase HPLC analyses were used to determine 15-LOX1/2 expression and their arachidonic acid metabolites in HRMVECs. The role of MEK1 in 15(S)-HETE-induced angiogenesis was studied using HRMVEC migration, tube formation, and basement membrane matrix plug angiogenesis. RESULTS: HRMVECs expressed both 15-LOX1 and 15-LOX2. Hypoxia induced the expression of 15-LOX1 and the production of its arachidonic acid metabolites 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) and 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE). 15(S)-HETE stimulated HRMVEC migration and tube formation as potently as 20 ng/mL fibroblast growth factor-2 (FGF-2). In addition, 15(S)-HETE stimulated the phosphorylation of ERK1/2, JNK1, p38 MAPK, and MEK1 in a time-dependent manner in these cells. Inhibition of MEK1 by pharmacologic and dominant-negative mutant approaches attenuated 15(S)-HETE-induced phosphorylation of ERK1/2 and JNK1 but not p38 MAPK. Blockade of ERK1/2 and JNK1 activation suppressed 15(S)-HETE-induced HRMVEC migration and tube formation and basement membrane matrix plug angiogenesis. Inhibition of p38 MAPK attenuated 15(S)-HETE-induced HRMVEC migration only. Inhibition of MEK1 also blocked 15(S)-HETE-induced HRMVEC migration and tube formation and basement membrane matrix plug angiogenesis. CONCLUSIONS: These results suggest that hypoxia, through the induction of 15-LOX1 expression, leads to the production of 15(S)-HETE in HRMVECs. In addition, 15(S)-HETE, through MEK1-dependent activation of ERK1/2 and JNK1, stimulates the angiogenic differentiation of HRMVECs and basement membrane matrix plug angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Hipoxia/metabolismo , MAP Quinasa Quinasa 1/fisiología , Neovascularización Retiniana/metabolismo , Vasos Retinianos/citología , Adenoviridae/genética , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Membrana Basal , Movimiento Celular , Células Cultivadas , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/farmacología , Vectores Genéticos , Humanos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
BMC Biol ; 4: 14, 2006 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-16672052

RESUMEN

BACKGROUND: The RB1 gene was the first tumor suppressor gene cloned from humans by studying genetic lesions in families with retinoblastoma. Children who inherit one defective copy of the RB1 gene have an increased susceptibility to retinoblastoma. Several years after the identification of the human RB1 gene, a targeted deletion of Rb was generated in mice. Mice with one defective copy of the Rb gene do not develop retinoblastoma. In this manuscript, we explore the different roles of the Rb family in human and mouse retinal development in order to better understand the species-specific difference in retinoblastoma susceptibility. RESULTS: We found that the Rb family of proteins (Rb, p107 and p130) are expressed in a dynamic manner during mouse retinal development. The primary Rb family member expressed in proliferating embryonic retinal progenitor cells in mice is p107, which is required for appropriate cell cycle exit during retinogenesis. The primary Rb family member expressed in proliferating postnatal retinal progenitor cells is Rb. p130 protein is expressed redundantly with Rb in postmitotic cells of the inner nuclear layer and the ganglion cell layer of the mouse retina. When Rb is inactivated in an acute or chronic manner during mouse retinal development, p107 is upregulated in a compensatory manner. Similarly, when p107 is inactivated in the mouse retina, Rb is upregulated. No changes in p130 expression were seen when p107, Rb or both were inactivated in the developing mouse retina. In the human retina, RB1 was the primary family member expressed throughout development. There was very little if any p107 expressed in the developing human retina. In contrast to the developing mouse retina, when RB1 was acutely inactivated in the developing human fetal retina, p107 was not upregulated in a compensatory manner. CONCLUSION: We propose that intrinsic genetic compensation between Rb and p107 prevents retinoblastoma in Rb- or p107-deficient mice, but this compensation does not occur in humans. Together, these data suggest a model that explains why humans are susceptible to retinoblastoma following RB1 loss, but mice require both Rb and p107 gene inactivation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes Supresores de Tumor , Retina/embriología , Proteína de Retinoblastoma/fisiología , Animales , Proliferación Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Especificidad de la Especie , Células Madre
13.
Neuron ; 94(3): 550-568.e10, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28472656

RESUMEN

In the developing retina, multipotent neural progenitors undergo unidirectional differentiation in a precise spatiotemporal order. Here we profile the epigenetic and transcriptional changes that occur during retinogenesis in mice and humans. Although some progenitor genes and cell cycle genes were epigenetically silenced during retinogenesis, the most dramatic change was derepression of cell-type-specific differentiation programs. We identified developmental-stage-specific super-enhancers and showed that most epigenetic changes are conserved in humans and mice. To determine how the epigenome changes during tumorigenesis and reprogramming, we performed integrated epigenetic analysis of murine and human retinoblastomas and induced pluripotent stem cells (iPSCs) derived from murine rod photoreceptors. The retinoblastoma epigenome mapped to the developmental stage when retinal progenitors switch from neurogenic to terminal patterns of cell division. The epigenome of retinoblastomas was more similar to that of the normal retina than that of retina-derived iPSCs, and we identified retina-specific epigenetic memory.


Asunto(s)
Carcinogénesis/genética , Diferenciación Celular/genética , Reprogramación Celular/genética , Metilación de ADN/genética , Epigénesis Genética , Código de Histonas/genética , Retina/metabolismo , Retinoblastoma/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Retina/embriología , Células Fotorreceptoras Retinianas Bastones/citología , Proteína de Retinoblastoma/genética
14.
J Comp Neurol ; 498(1): 112-28, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16856163

RESUMEN

The retinoblastoma gene (Rb) regulates neural progenitor cell proliferation and cell fate specification and differentiation. For the developing mouse retina, two distinct functions of Rb have been described: regulation of retinal progenitor cell proliferation and rod photoreceptor development. Cells that would normally become rods fail to mature and remain as immature cells in the outer nuclear layer in the adult. By using Chx10-Cre;Rb(Lox/-) mice, we generated a chimeric retina with alternating apical-basal stripes of wild-type and Rb-deficient tissue. This provides a unique model with which to study synaptogenesis at the outer plexiform layer within regions that lack differentiated rods. In regions where rods failed to differentiate, the outer plexiform layer (OPL) was disrupted. Horizontal cells formed, and their somata were appropriately aligned, but their neurites did not project laterally. Instead many horizonal cell neurites extended apically, forming ectopic synapses with photoreceptors at all levels of the outer nuclear layer. These ectopic photoreceptor terminals contained synaptic ribbons, horizontal cell processes with synaptic vesicles, and a single mitochrondrion characteristic of rod spherules. Rb-deficient bipolar cells differentiated normally, extended dendrites to the OPL, and formed synapses that were indistinguishable from adjacent wild-type cells. In contrast to OPL-positioned synapses, ectopic synapses did not contain bipolar dendrites. This finding suggests that horizontal cells and photoreceptors can form stable synapses that are devoid of bipolar dendrites outside the normal boundaries of the OPL. Finally, analysis of P4, P7, P12, and P15 retinae suggests that the apical horizontal cell processes result from their failure to establish their normal lateral projections during development.


Asunto(s)
Diferenciación Celular/genética , Vías Nerviosas/anomalías , Degeneración Retiniana/genética , Células Fotorreceptoras Retinianas Bastones/anomalías , Proteína de Retinoblastoma/genética , Sinapsis/patología , Animales , Animales Recién Nacidos , Quimera/anomalías , Quimera/crecimiento & desarrollo , Coristoma/genética , Coristoma/metabolismo , Coristoma/patología , Dendritas/patología , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Eliminación de Gen , Ratones , Ratones Noqueados , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/ultraestructura , Terminales Presinápticos/patología , Terminales Presinápticos/ultraestructura , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/ultraestructura , Degeneración Retiniana/metabolismo , Degeneración Retiniana/fisiopatología , Células Fotorreceptoras Retinianas Bastones/crecimiento & desarrollo , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Células Madre/citología , Células Madre/metabolismo , Sinapsis/ultraestructura
15.
BMC Ophthalmol ; 5: 25, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16262907

RESUMEN

BACKGROUND: Retinal pigment epithelium cells play an important role in the pathogenesis of age related macular degeneration. Their morphological, molecular and functional phenotype changes in response to various stresses. Functional profiling of genes can provide useful information about the physiological state of cells and how this state changes in response to disease or treatment. In this study, we have constructed a functional profile of the genes expressed by the ARPE-19 cell line of retinal pigment epithelium. METHODS: Using Affymetrix MAS 5.0 microarray analysis, genes expressed by ARPE-19 cells were identified. Using GeneChip annotations, these genes were classified according to their known functions to generate a functional gene expression profile. RESULTS: We have determined that of approximately 19,044 unique gene sequences represented on the HG-U133A GeneChip, 6,438 were expressed in ARPE-19 cells irrespective of the substrate on which they were grown (plastic, fibronectin, collagen, or Matrigel). Rather than focus our subsequent analysis on the identity or level of expression of each individual gene in this large data set, we examined the number of genes expressed within 130 functional categories. These categories were selected from a library of HG-U133A GeneChip annotations linked to the Affymetrix MAS 5.0 data sets. Using this functional classification scheme, we were able to categorize about 70% of the expressed genes and condense the original data set of over 6,000 data points into a format with 130 data points. The resulting ARPE-19 Functional Gene Expression Profile is displayed as a percentage of ARPE-19-expressed genes. CONCLUSION: The Profile can readily be compared with equivalent microarray data from other appropriate samples in order to highlight cell-specific attributes or treatment-induced changes in gene expression. The usefulness of these analyses is based on the assumption that the numbers of genes expressed within a functional category provide an indicator of the overall level of activity within that particular functional pathway.


Asunto(s)
Proteínas del Ojo/genética , Perfilación de la Expresión Génica , Expresión Génica/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Epitelio Pigmentado Ocular/metabolismo , Línea Celular , Humanos , Sensibilidad y Especificidad
16.
Cell Stem Cell ; 17(1): 101-15, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26140606

RESUMEN

Cell-based therapies to treat retinal degeneration are now being tested in clinical trials. However, it is not known whether the source of stem cells is important for the production of differentiated cells suitable for transplantation. To test this, we generated induced pluripotent stem cells (iPSCs) from murine rod photoreceptors (r-iPSCs) and scored their ability to make retinae by using a standardized quantitative protocol called STEM-RET. We discovered that r-iPSCs more efficiently produced differentiated retinae than did embryonic stem cells (ESCs) or fibroblast-derived iPSCs (f-iPSCs). Retinae derived from f-iPSCs had fewer amacrine cells and other inner nuclear layer cells. Integrated epigenetic analysis showed that DNA methylation contributes to the defects in f-iPSC retinogenesis and that rod-specific CTCF insulator protein-binding sites may promote r-iPSC retinogenesis. Together, our data suggest that the source of stem cells is important for producing retinal neurons in three-dimensional (3D) organ cultures.


Asunto(s)
Epigénesis Genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Retina/crecimiento & desarrollo , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Línea Celular , Reprogramación Celular , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Ratones Transgénicos , Retina/citología , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Degeneración Retiniana/terapia
17.
Invest Ophthalmol Vis Sci ; 43(9): 3081-90, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12202533

RESUMEN

PURPOSE: To measure the concentrations of polyamines, determine their cellular and subcellular localization, and analyze effects of their depletion in developing rabbit retina. METHODS: Isolated retinas at different developmental stages were analyzed for polyamine content by high-performance liquid chromatography (HPLC). An antibody against polyamines was used to localize endogenous stores in both freshly harvested retinas and neonatal retinal explants. To determine the effects of polyamine depletion on immature retina, neonatal explants were cultured in the presence or absence of alpha-difluoromethylornithine (DFMO), an inhibitor of the polyamine synthetic enzyme ornithine decarboxylase (ODC). Similar studies were also performed on dissociated cell cultures. Tissue was assessed using standard histologic stains as well as cell-specific markers (peanut agglutinin for cone photoreceptors and calbindin for horizontal cells). RESULTS: Retinal polyamine content was highest at birth, remained relatively high during the first postnatal week, and then steadily decreased to adult levels. At all ages analyzed, spermine concentration was higher than putrescine or spermidine; however, the differential was greatest in the adult. Polyamine immunoreactivity was localized to distal processes of both rods and cones during development. Strong immunoreactivity was maintained in adult cone inner and outer segments; comparatively weak staining was observed in the adult rods. Heavy staining of ganglion cells was present throughout development but was localized in the cytoplasm in immature cells and in the nucleus in the adult. Amacrine cells stained only in the adult. Polyamine depletion caused a disruption of immature cones, evident in the loss of their somata in the outer nuclear layer, in their processes in the outer plexiform layer in retinal explants, and in their decreased association with horizontal cells in dissociated cell culture. CONCLUSIONS: The relatively high concentrations of polyamines in neonatal retina and their discrete localization in developing photoreceptor outer segments and ganglion cells suggests an important role for these compounds in development. The disruption of cone-specific markers in polyamine-depleted retinas indicates a specific reliance on polyamines for expression of normal cone morphology or morphologic development. These developmental effects may involve polyamine-sensitive ion channels, which are known to exist in retina, or direct interactions with specialized cytoskeletal elements within outer segments.


Asunto(s)
Poliaminas Biogénicas/fisiología , Células Fotorreceptoras Retinianas Conos/crecimiento & desarrollo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Poliaminas Biogénicas/análisis , Calbindinas , Técnicas de Cultivo de Célula , Cromatografía Líquida de Alta Presión , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Técnicas para Inmunoenzimas , Inhibidores de la Ornitina Descarboxilasa , Conejos , Retina/química , Retina/efectos de los fármacos , Retina/crecimiento & desarrollo , Células Fotorreceptoras Retinianas Conos/química , Proteína G de Unión al Calcio S100/metabolismo , Fracciones Subcelulares
18.
Invest Ophthalmol Vis Sci ; 43(4): 1228-33, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11923270

RESUMEN

PURPOSE: Migration of retinal pigment epithelial (RPE) cells can be triggered by disruption of the RPE monolayer or injury to the neural retina. Migrating cells may re-establish a confluent monolayer, or they may invade the neural retina and disrupt visual function. The purpose of this study was to examine the role of endogenous polyamines in mechanisms of RPE migration. METHODS: Endogenous polyamine levels were determined in an immortalized RPE cell line, D407, using HPLC. Activities of the two rate-limiting enzymes for polyamine synthesis, ornithine decarboxylase (ODC), and S-adenosylmethionine decarboxylase (SAMdc), were measured by liberation of ((14)CO(2))(.) Migration was assessed in confluent cultures by determining the number of cells migrating into a mechanically denuded area. All measurements were obtained both in control cultures and in cultures treated with synthesis inhibitors that deplete endogenous polyamines. Subcellular localization of endogenous polyamines was determined using a polyamine antibody. RESULTS: The polyamines, spermidine and spermine, as well as their precursor, putrescine, were normal constituents of RPE cells. The two rate-limiting synthetic enzymes were also present, and their activities were stimulated dramatically by addition of serum to the culture medium. Cell migration was similarly stimulated by serum exposure. When endogenous polyamines were depleted, migration was blocked. When polyamines were replenished through uptake, migration was restored. Polyamine immunoreactivity was limited to membrane patches in quiescent cells. In actively migrating and dividing cells, immunoreactivity was enhanced throughout the cytoplasm. CONCLUSIONS: Polyamines are essential for RPE migration. Pharmacologic manipulation of the polyamine pathway could provide a therapeutic strategy for regulating anomalous migration.


Asunto(s)
Movimiento Celular/fisiología , Mitoguazona/análogos & derivados , Epitelio Pigmentado Ocular/citología , Putrescina/fisiología , Espermidina/fisiología , Espermina/fisiología , Adenosilmetionina Descarboxilasa/antagonistas & inhibidores , Adenosilmetionina Descarboxilasa/metabolismo , Línea Celular , Cromatografía Líquida de Alta Presión , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Mitoguazona/farmacología , Ornitina Descarboxilasa/metabolismo , Inhibidores de la Ornitina Descarboxilasa , Epitelio Pigmentado Ocular/enzimología
19.
Brain Res Dev Brain Res ; 145(1): 93-105, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-14519497

RESUMEN

Mice represent a valuable species for studies of development and disease. With the availability of transgenic models for retinal degeneration in this species, information regarding development and structure of mouse retina has become increasingly important. Of special interest is the differentiation and synaptogenesis of photoreceptors since these cells are predominantly involved in hereditary retinal degenerations. Thus, some of the keys to future clinical management of these retinal diseases may lie in understanding the molecular mechanisms of outer retinal development. In this study, we describe the expression of markers for photoreceptors (recoverin), horizontal cells (calbindin), bipolar cells (protein kinase C; PKC) and cytoskeletal elements pivotal to axonogenesis (beta-tubulin and actin) during perinatal development of mouse retina. Immunocytochemical localization of recoverin, calbindin, PKC and beta-tubulin was monitored in developing mouse retina (embryonic day (E) 18.5 to postnatal day (PN) 14), whereas f-actin was localized by Phalloidin binding. Recoverin immunoreactive cells, presumably the photoreceptors, were observed embryonically (E 18.5) and their number increased until PN 14. Neurite projections from the immunoreactive cells towards the outer plexiform layer (OPL) were noted at PN 0 and these processes reached the OPL at PN 7 coincident with histological evidence for the differentiation of the OPL. Outer segments, all the cell bodies in the ONL, as well as the OPL were immunoreactive to recoverin at PN 14. Calbindin immunoreactive horizontal cells were also present in E 18.5 retinas. These cells became progressively displaced proximally as the ONL developed. A calbindin immunoreactive plexus was seen in the OPL at PN 7. PKC immunoreactive bipolar cells developed postnatally, becoming distinguished at PN 7. Both beta-tubulin and actin immunoreactive cells were present in the IPL as early as E 18.5; however, appearance of processes labeled with these markers in the OPL was delayed until PN 7, concurrent with the first appearance of photoreceptor neurites, development of the horizontal cell plexus, and development of synaptophysin immunoreactivity at this location. These results provide a developmental timeframe for the expression of recoverin, calbindin, synaptophysin, beta-tubulin and actin. Our findings suggest that the time between PN 3 and PN 7 represents a critical period during which elements of the OPL are assembled.


Asunto(s)
Proteínas del Ojo , Lipoproteínas , Proteínas del Tejido Nervioso , Retina/crecimiento & desarrollo , Envejecimiento , Animales , Animales Recién Nacidos , Calbindinas , Proteínas de Unión al Calcio/metabolismo , Embrión de Mamíferos , Femenino , Hipocalcina , Inmunohistoquímica , Masculino , Ratones , Faloidina/farmacocinética , Proteína Quinasa C/metabolismo , Recoverina , Retina/citología , Retina/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Sinaptofisina/metabolismo , Tubulina (Proteína)/metabolismo
20.
Oncotarget ; 5(20): 9594-608, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25338120

RESUMEN

The retinoblastoma (Rb) family of proteins are key regulators of cell cycle exit during development and their deregulation is associated with cancer. Rb is critical for normal retinal development and germline mutations lead to retinoblastoma making retinae an attractive system to study Rb family signaling. Rb coordinates proliferation and differentiation through the E2f family of transcription factors, a critical interaction for the role of Rb in retinal development and tumorigenesis. However, whether the roles of the different E2fs are interchangeable in controlling development and tumorigenesis in the retina or if they have selective functions remains unknown. In this study, we found that E2f family members play distinct roles in the development and tumorigenesis. In Rb;p107-deficient retinae, E2f1 and E2f3 inactivation rescued tumor formation but only E2f1 rescued the retinal development phenotype. This allowed the identification of key target genes for Rb/E2f family signaling contributing to tumorigenesis and those contributing to developmental defects. We found that Sox4 and Sox11 genes contribute to the developmental phenotype and Hells and Uhrf1 contribute to tumorigenesis. Using orthotopic human xenografts, we validated that upregulation of HELLS and UHRF1 is essential for the tumor phenotype. Also, these epigenetic regulators are important for the regulation of SYK.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Cromatina/genética , ADN Helicasas/genética , Neoplasias de la Retina/genética , Retinoblastoma/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Cromatina/metabolismo , ADN Helicasas/metabolismo , Progresión de la Enfermedad , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F3/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/metabolismo , Retinoblastoma/patología , Quinasa Syk , Transfección , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA