Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Development ; 145(9)2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29650591

RESUMO

Precise control of the relative ratio of retinal neurons and glia generated during development is essential for visual function. We show that Lhx2, which encodes a LIM-homeodomain transcription factor essential for specification and differentiation of retinal Müller glia, also plays a crucial role in the development of retinal neurons. Overexpression of Lhx2 with its transcriptional co-activator Ldb1 triggers cell cycle exit and inhibits both Notch signaling and retinal gliogenesis. Lhx2/Ldb1 overexpression also induces the formation of wide-field amacrine cells (wfACs). In contrast, Rnf12, which encodes a negative regulator of LDB1, is necessary for the initiation of retinal gliogenesis. We also show that Lhx2-dependent neurogenesis and wfAC formation requires Ascl1 and Neurog2, and that Lhx2 is necessary for their expression, although overexpression of Lhx2/Ldb1 does not elevate expression of these proneural bHLH factors. Finally, we demonstrate that the relative level of the LHX2-LDB1 complex in the retina decreases in tandem with the onset of gliogenesis. These findings show that control of Lhx2 function by Ldb1 and Rnf12 underpins the coordinated differentiation of neurons and Müller glia in postnatal retina.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Ependimogliais/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Neurogênese/fisiologia , Neurônios Retinianos/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/genética , Células Ependimogliais/citologia , Proteínas com Domínio LIM/genética , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios Retinianos/citologia , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética
2.
Development ; 143(21): 3994-4002, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27633990

RESUMO

Fibroblast growth factor (FGF) signaling is an essential regulator of lens epithelial cell proliferation and survival, as well as lens fiber cell differentiation. However, the identities of these FGF factors, their source tissue and the genes that regulate their synthesis are unknown. We have found that Chx10-Cre;Lhx2lox/lox mice, which selectively lack Lhx2 expression in neuroretina from E10.5, showed an early arrest in lens fiber development along with severe microphthalmia. These mutant animals showed reduced expression of multiple neuroretina-expressed FGFs and canonical FGF-regulated genes in neuroretina. When FGF expression was genetically restored in Lhx2-deficient neuroretina of Chx10-Cre;Lhx2lox/lox mice, we observed a partial but nonetheless substantial rescue of the defects in lens cell proliferation, survival and fiber differentiation. These data demonstrate that neuroretinal expression of Lhx2 and neuroretina-derived FGF factors are crucial for lens fiber development in vivo.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Proteínas com Homeodomínio LIM/fisiologia , Cristalino/embriologia , Organogênese/genética , Neurônios Retinianos/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Embrião de Mamíferos , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/genética , Cristalino/metabolismo , Camundongos , Camundongos Transgênicos , Microftalmia/embriologia , Microftalmia/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Neurônios Retinianos/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética
3.
J Neurosci ; 36(8): 2391-405, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26911688

RESUMO

Müller glia (MG) are the only glial cell type produced by the neuroepithelial progenitor cells that generate the vertebrate retina. MG are required to maintain retinal homeostasis and support the survival of retinal neurons. Furthermore, in certain vertebrate classes, MG function as adult stem cells, mediating retinal regeneration in response to injury. However, the mechanisms that regulate MG development are poorly understood because there is considerable overlap in gene expression between retinal progenitor cells and differentiated MG. We show that the LIM homeodomain transcription factor Lhx2 is required for the development of MG in the mouse retina. Temporally controlled knock-out studies reveal a requirement for Lhx2 during all stages of MG development, ranging from the proliferation of gliocompetent retinal progenitors, activation of Müller-specific gene expression, and terminal differentiation of MG morphological features. We show that Lhx2 regulates gliogenesis in part by regulating directly the expression of Notch pathway genes including Notch1, Dll1, and Dll3 and gliogenic transcription factors such as Hes1, Hes5, Sox8, and Rax. Conditional knock-out of Lhx2 resulted in a rapid downregulation of Notch pathway genes and loss of Notch signaling. We further demonstrate that Müller gliogenesis induced by misexpression of the potently gliogenic Notch pathway transcriptional effector Hes5 requires Lhx2 expression. These results indicate that Lhx2 not only directly regulates expression of Notch signaling pathway components, but also acts together with the gliogenic Notch pathway to drive MG specification and differentiation.


Assuntos
Proteínas com Homeodomínio LIM/biossíntese , Neuroglia/metabolismo , Receptor Notch1/biossíntese , Neurônios Retinianos/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/biossíntese , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Retina
4.
BMC Bioinformatics ; 18(1): 355, 2017 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-28750606

RESUMO

BACKGROUND: Computational prediction of transcription factor (TF) binding sites in different cell types is challenging. Recent technology development allows us to determine the genome-wide chromatin accessibility in various cellular and developmental contexts. The chromatin accessibility profiles provide useful information in prediction of TF binding events in various physiological conditions. Furthermore, ChIP-Seq analysis was used to determine genome-wide binding sites for a range of different TFs in multiple cell types. Integration of these two types of genomic information can improve the prediction of TF binding events. RESULTS: We assessed to what extent a model built upon on other TFs and/or other cell types could be used to predict the binding sites of TFs of interest. A random forest model was built using a set of cell type-independent features such as specific sequences recognized by the TFs and evolutionary conservation, as well as cell type-specific features derived from chromatin accessibility data. Our analysis suggested that the models learned from other TFs and/or cell lines performed almost as well as the model learned from the target TF in the cell type of interest. Interestingly, models based on multiple TFs performed better than single-TF models. Finally, we proposed a universal model, BPAC, which was generated using ChIP-Seq data from multiple TFs in various cell types. CONCLUSION: Integrating chromatin accessibility information with sequence information improves prediction of TF binding.The prediction of TF binding is transferable across TFs and/or cell lines suggesting there are a set of universal "rules". A computational tool was developed to predict TF binding sites based on the universal "rules".


Assuntos
Cromatina/metabolismo , Modelos Genéticos , Fatores de Transcrição/metabolismo , Algoritmos , Área Sob a Curva , Sítios de Ligação , Linhagem Celular Tumoral , Cromatina/química , Montagem e Desmontagem da Cromatina , DNA/química , DNA/metabolismo , Humanos , Ligação Proteica , Curva ROC , Fatores de Transcrição/química
5.
J Neurosci ; 34(50): 16809-20, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25505333

RESUMO

Hypothalamic tanycytes, a radial glial-like ependymal cell population that expresses numerous genes selectively enriched in embryonic hypothalamic progenitors and adult neural stem cells, have recently been observed to serve as a source of adult-born neurons in the mammalian brain. The genetic mechanisms that regulate the specification and maintenance of tanycyte identity are unknown, but are critical for understanding how these cells can act as adult neural progenitor cells. We observe that LIM (Lin-11, Isl-1, Mec-3)-homeodomain gene Lhx2 is selectively expressed in hypothalamic progenitor cells and tanycytes. To test the function of Lhx2 in tanycyte development, we used an intersectional genetic strategy to conditionally delete Lhx2 in posteroventral hypothalamic neuroepithelium, both embryonically and postnatally. We observed that tanycyte development was severely disrupted when Lhx2 function was ablated during embryonic development. Lhx2-deficient tanycytes lost expression of tanycyte-specific genes, such as Rax, while also displaying ectopic expression of genes specific to cuboid ependymal cells, such as Rarres2. Ultrastructural analysis revealed that mutant tanycytes exhibited a hybrid identity, retaining radial morphology while becoming multiciliated. In contrast, postnatal loss of function of Lhx2 resulted only in loss of expression of tanycyte-specific genes. Using chromatin immunoprecipitation, we further showed that Lhx2 directly regulated expression of Rax, an essential homeodomain factor for tanycyte development. This study identifies Lhx2 as a key intrinsic regulator of tanycyte differentiation, sustaining Rax-dependent activation of tanycyte-specific genes while also inhibiting expression of ependymal cell-specific genes. These findings provide key insights into the transcriptional regulatory network specifying this still poorly characterized cell type.


Assuntos
Diferenciação Celular/fisiologia , Células Ependimogliais/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Neurogênese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos
6.
Proc Natl Acad Sci U S A ; 109(12): 4657-62, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393024

RESUMO

Müller glia are the primary glial subtype in the retina and perform a wide range of physiological tasks in support of retinal function, but little is known about the transcriptional network that maintains these cells in their differentiated state. We report that selective deletion of the LIM homeodomain transcription factor Lhx2 from mature Müller glia leads to the induction of reactive retinal gliosis in the absence of injury. Furthermore, Lhx2 expression is also down-regulated in Prph2(Rd2/Rd2) animals immediately before the onset of reactive gliosis. Analysis of conditional Lhx2 knockouts showed that gliosis was hypertrophic but not proliferative. Aging of experimental animals demonstrated that constitutive reactive gliosis induced by deletion of Lhx2 reduced rates of ongoing apoptosis and compromised both rod and cone photoreceptor function. Additionally, these animals showed a dramatically reduced ability to induce expression of secreted neuroprotective factors and displayed enhanced rates of apoptosis in light-damage assays. We provide in vivo evidence that Lhx2 actively maintains mature Müller glia in a nonreactive state, with loss of function initiating a specific program of nonproliferative hypertrophic gliosis.


Assuntos
Gliose/patologia , Proteínas com Homeodomínio LIM/fisiologia , Retina/patologia , Fatores de Transcrição/fisiologia , Animais , Antineoplásicos Hormonais/farmacologia , Regulação da Expressão Gênica , Gliose/genética , Proteínas com Homeodomínio LIM/genética , Luz , Camundongos , Camundongos Knockout , Modelos Biológicos , Doenças Neurodegenerativas/metabolismo , Neuroglia/fisiologia , Estrutura Terciária de Proteína , Retina/fisiologia , Tamoxifeno/farmacologia , Fatores de Transcrição/genética
7.
Cells ; 11(5)2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35269428

RESUMO

Retinal neurogenesis is driven by concerted actions of transcription factors, some of which are expressed in a continuum and across several cell subtypes throughout development. While seemingly redundant, many factors diversify their regulatory outcome on gene expression, by coordinating variations in chromatin landscapes to drive divergent retinal specification programs. Recent studies have furthered the understanding of the epigenetic contribution to the progression of age-related macular degeneration, a leading cause of blindness in the elderly. The knowledge of the epigenomic mechanisms that control the acquisition and stabilization of retinal cell fates and are evoked upon damage, holds the potential for the treatment of retinal degeneration. Herein, this review presents the state-of-the-art approaches to investigate the retinal epigenome during development, disease, and reprogramming. A pipeline is then reviewed to functionally interrogate the epigenetic and transcriptional networks underlying cell fate specification, relying on a truly unbiased screening of open chromatin states. The related work proposes an inferential model to identify gene regulatory networks, features the first footprinting analysis and the first tentative, systematic query of candidate pioneer factors in the retina ever conducted in any model organism, leading to the identification of previously uncharacterized master regulators of retinal cell identity, such as the nuclear factor I, NFI. This pipeline is virtually applicable to the study of genetic programs and candidate pioneer factors in any developmental context. Finally, challenges and limitations intrinsic to the current next-generation sequencing techniques are discussed, as well as recent advances in super-resolution imaging, enabling spatio-temporal resolution of the genome.


Assuntos
Epigenoma , Fatores de Transcrição NFI , Idoso , Cromatina/genética , Epigenoma/genética , Humanos , Fatores de Transcrição NFI/genética , Neurogênese/genética , Retina
8.
J Neurosci ; 30(7): 2521-32, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20164337

RESUMO

A variety of chromatin remodeling complexes are thought to orchestrate transcriptional programs that lead neuronal precursors from earliest commitment to terminal differentiation. Here we show that mammalian neurons have a specialized chromatin remodeling enzyme arising from a neurospecific splice variant of LSD1/KDM1, histone lysine specific demethylase 1, whose demethylase activity on Lys4 of histone H3 has been related to gene repression. We found that alternative splicing of LSD1 transcript generates four full-length isoforms from combinatorial retention of two identified exons: the 4 aa exon E8a is internal to the amine oxidase domain, and its inclusion is restricted to the nervous system. Remarkably, the expression of LSD1 splice variants is dynamically regulated throughout cortical development, particularly during perinatal stages, with a progressive increase of LSD1 neurospecific isoforms over the ubiquitous ones. Notably, the same LSD1 splice dynamics can be fairly recapitulated in cultured cortical neurons. Functionally, LSD1 isoforms display in vitro a comparable demethylase activity, yet the inclusion of the sole exon E8a reduces LSD1 repressor activity on a reporter gene. Additional distinction among isoforms is supported by the knockdown of neurospecific variants in cortical neurons resulting in the inhibition of neurite maturation, whereas overexpression of the same variants enhances it. Instead, perturbation of LSD1 isoforms that are devoid of the neurospecific exon elicits no morphogenic effect. Collectively, results demonstrate that the arousal of neuronal LSD1 isoforms pacemakes early neurite morphogenesis, conferring a neurospecific function to LSD1 epigenetic activity.


Assuntos
Processamento Alternativo/genética , Histona Desmetilases/genética , Morfogênese/genética , Neuritos/fisiologia , Neurônios/citologia , Animais , Encéfalo/citologia , Células Cultivadas , Embrião de Mamíferos , Éxons/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genoma , Proteínas de Fluorescência Verde/genética , Guanilato Quinases/metabolismo , Células HeLa , Humanos , Imunoprecipitação/métodos , Modelos Moleculares , Proteínas do Tecido Nervoso/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Receptores de Glutamato/metabolismo , Proteínas Associadas SAP90-PSD95 , Transfecção/métodos
9.
J Neurosci ; 30(17): 5830-42, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20427644

RESUMO

Activity-dependent changes in synaptic structure and spine morphology are required for learning and memory, and depend on protein translation. We show that the kinase for eukaryotic elongation factor 2 (eEF2K) regulates dendritic spine stability and synaptic structure by modulating activity-dependent dendritic BDNF synthesis. Specifically RNAi knockdown of eEF2K reduces dendritic spine stability and inhibits dendritic BDNF protein expression; whereas overexpression of a constitutively activated eEF2K induces spine maturation and increases expression of dendritic BDNF. Furthermore, BDNF overexpression rescues the spine stability reduced by RNAi knockdown of eEF2K. We also show that synaptic activity-dependent spine maturation and dendritic BDNF protein expression depend on mGluR/EF2K-induced eEF2 phosphorylation. We propose that the eEF2K/eEF2 pathway is a key biochemical sensor that couple neuronal activity to spine plasticity, by controlling the dendritic translation of BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Espinhas Dendríticas/fisiologia , Hipocampo/fisiologia , Fator 2 de Elongação de Peptídeos/metabolismo , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Células Cultivadas , Espinhas Dendríticas/enzimologia , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Técnicas de Silenciamento de Genes , Técnicas In Vitro , Neurônios/fisiologia , Fosforilação , Interferência de RNA , Ratos , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais , Sinapses/enzimologia
10.
Commun Biol ; 2: 142, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31044167

RESUMO

Retinal neurogenesis occurs through partially overlapping temporal windows, driven by concerted actions of transcription factors which, in turn, may contribute to the establishment of divergent genetic programs in the developing retina by coordinating variations in chromatin landscapes. Here we comprehensively profile murine retinal progenitors by integrating next generation sequencing methods and interrogate changes in chromatin accessibility at embryonic and post-natal stages. An unbiased search for motifs in open chromatin regions identifies putative factors involved in the developmental progression of the epigenome in retinal progenitor cells. Among these factors, the transcription factor LHX2 exhibits a developmentally regulated cis-regulatory repertoire and stage-dependent motif instances. Using loss-of-function assays, we determine LHX2 coordinates variations in chromatin accessibility, by competition for nucleosome occupancy and secondary regulation of candidate pioneer factors.


Assuntos
Epigenômica/métodos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas com Homeodomínio LIM/metabolismo , Neurogênese/genética , Retina/embriologia , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Cromatina/metabolismo , Sequenciamento de Cromatina por Imunoprecipitação , Feminino , Técnicas de Inativação de Genes , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Camundongos Knockout , Nucleossomos/metabolismo , RNA-Seq , Fatores de Transcrição/genética
11.
Cell Syst ; 8(5): 395-411.e8, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31121116

RESUMO

Analysis of gene expression in single cells allows for decomposition of cellular states as low-dimensional latent spaces. However, the interpretation and validation of these spaces remains a challenge. Here, we present scCoGAPS, which defines latent spaces from a source single-cell RNA-sequencing (scRNA-seq) dataset, and projectR, which evaluates these latent spaces in independent target datasets via transfer learning. Application of developing mouse retina to scRNA-Seq reveals intrinsic relationships across biological contexts and assays while avoiding batch effects and other technical features. We compare the dimensions learned in this source dataset to adult mouse retina, a time-course of human retinal development, select scRNA-seq datasets from developing brain, chromatin accessibility data, and a murine-cell type atlas to identify shared biological features. These tools lay the groundwork for exploratory analysis of scRNA-seq data via latent space representations, enabling a shift in how we compare and identify cells beyond reliance on marker genes or ensemble molecular identity.


Assuntos
Perfilação da Expressão Gênica/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Algoritmos , Animais , Bases de Dados Genéticas , Feminino , Humanos , Aprendizado de Máquina , Masculino , Camundongos , Camundongos Transgênicos , Retina/embriologia , Software , Transcriptoma/genética , Sequenciamento do Exoma/métodos
12.
Nat Commun ; 9(1): 1364, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636475

RESUMO

Age-related macular degeneration (AMD) is a significant cause of vision loss in the elderly. The extent to which epigenetic changes regulate AMD progression is unclear. Here we globally profile chromatin accessibility using ATAC-Seq in the retina and retinal pigmented epithelium (RPE) from AMD and control patients. Global decreases in chromatin accessibility occur in the RPE with early AMD, and in the retina of advanced disease, suggesting that dysfunction in the RPE drives disease onset. Footprints of photoreceptor and RPE-specific transcription factors are enriched in differentially accessible regions (DARs). Genes associated with DARs show altered expression in AMD. Cigarette smoke treatment of RPE cells recapitulates chromatin accessibility changes seen in AMD, providing an epigenetic link between a known risk factor for AMD and AMD pathology. Finally, overexpression of HDAC11 is partially responsible for the observed reduction in chromatin accessibility, suggesting that HDAC11 may be a potential new therapeutic target for AMD.


Assuntos
Cromatina/química , Epigênese Genética , Proteínas do Olho/genética , Histona Desacetilases/genética , Degeneração Macular/genética , Fatores de Transcrição/genética , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Cromatina/metabolismo , Misturas Complexas/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Histona Desacetilases/metabolismo , Humanos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Masculino , Cultura Primária de Células , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Fumaça/análise , Nicotiana/química , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA