Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 112022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36459481

RESUMO

An important question in organogenesis is how tissue-specific transcription factors interact with signaling pathways. In some cases, transcription factors define the context for how signaling pathways elicit tissue- or cell-specific responses, and in others, they influence signaling through transcriptional regulation of signaling components or accessory factors. We previously showed that during optic vesicle patterning, the Lim-homeodomain transcription factor Lhx2 has a contextual role by linking the Sonic Hedgehog (Shh) pathway to downstream targets without regulating the pathway itself. Here, we show that during early retinal neurogenesis in mice, Lhx2 is a multilevel regulator of Shh signaling. Specifically, Lhx2 acts cell autonomously to control the expression of pathway genes required for efficient activation and maintenance of signaling in retinal progenitor cells. The Shh co-receptors Cdon and Gas1 are candidate direct targets of Lhx2 that mediate pathway activation, whereas Lhx2 directly or indirectly promotes the expression of other pathway components important for activation and sustained signaling. We also provide genetic evidence suggesting that Lhx2 has a contextual role by linking the Shh pathway to downstream targets. Through these interactions, Lhx2 establishes the competence for Shh signaling in retinal progenitors and the context for the pathway to promote early retinal neurogenesis. The temporally distinct interactions between Lhx2 and the Shh pathway in retinal development illustrate how transcription factors and signaling pathways adapt to meet stage-dependent requirements of tissue formation.


Assuntos
Proteínas Hedgehog , Neurogênese , Camundongos , Animais , Neurogênese/genética , Retina , Transdução de Sinais , Fatores de Transcrição , Proteínas com Homeodomínio LIM/genética
2.
Development ; 146(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30936115

RESUMO

Murine cardiac and hematopoietic progenitors are derived from Mesp1+ mesoderm. Cdx function impacts both yolk sac hematopoiesis and cardiogenesis in zebrafish, suggesting that Cdx family members regulate early mesoderm cell fate decisions. We found that Cdx2 occupies a number of transcription factor loci during embryogenesis, including key regulators of both cardiac and blood development, and that Cdx function is required for normal expression of the cardiogenic transcription factors Nkx2-5 and Tbx5 Furthermore, Cdx and Brg1, an ATPase subunit of the SWI/SNF chromatin remodeling complex, co-occupy a number of loci, suggesting that Cdx family members regulate target gene expression through alterations in chromatin architecture. Consistent with this, we demonstrate loss of Brg1 occupancy and altered chromatin structure at several cardiogenic genes in Cdx-null mutants. Finally, we provide evidence for an onset of Cdx2 expression at E6.5 coinciding with egression of cardiac progenitors from the primitive streak. Together, these findings suggest that Cdx functions in multi-potential mesoderm to direct early cell fate decisions through transcriptional regulation of several novel target genes, and provide further insight into a potential epigenetic mechanism by which Cdx influences target gene expression.


Assuntos
Fator de Transcrição CDX2/metabolismo , Animais , Fator de Transcrição CDX2/genética , Imunoprecipitação da Cromatina , Feminino , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Masculino , Camundongos , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
3.
Bio Protoc ; 7(3)2017 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-28367479

RESUMO

All seven retinal cell types that make up the mature retina are generated from a common, multipotent pool of retinal progenitor cells (RPCs) (Wallace, 2011). One way that RPCs know when sufficient numbers of particular cell-types have been generated is through negative feedback signals, which are emitted by differentiated cells and must reach threshold levels to block additional differentiation of that cell type. A key assay to assess whether negative feedback signals are emitted by differentiated cells is a heterochronic pellet assay in which early stage RPCs are dissociated and labeled with BrdU, then mixed with a 20-fold excess of dissociated differentiated cells. The combined cells are then re-aggregated and cultured as a pellet on a membrane for 7-10 days in vitro. During this time frame, RPCs will differentiate, and the fate of the BrdU+ RPCs can be assessed using cell type-specific markers. Investigators who developed this pellet assay initially demonstrated that neonatal RPCs give rise to rods on an accelerated schedule compared to embryonic RPCs when the two cell types are mixed together (Watanabe and Raff, 1990; Watanabe et al., 1997). We have used this assay to demonstrate that sonic hedgehog (Shh), which we found acts as a negative regulator of retinal ganglion cell (RGC) differentiation, promotes RPC proliferation (Jensen and Wallace, 1997; Ringuette et al., 2014). More recently we modified the heterochronic pellet assay to assess the role of feedback signals for retinal amacrine cells, identifying transforming growth factor ß2 (Tgfß2) as a negative feedback signal, and Pten as a modulator of the Tgfß2 response (Ma et al., 2007; Tachibana et al., 2016). This assay can be adapted to other lineages and tissues to assess cell-cell interactions between two different cell-types (heterotypic) in either an isochronic or heterochronic manner.

4.
Dev Biol ; 422(2): 115-124, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28065741

RESUMO

The Cdx transcription factors play essential roles in primitive hematopoiesis in the zebrafish where they exert their effects, in part, through regulation of hox genes. Defects in hematopoiesis have also been reported in Cdx mutant murine embryonic stem cell models, however, to date no mouse model reflecting the zebrafish Cdx mutant hematopoietic phenotype has been described. This is likely due, in part, to functional redundancy among Cdx members and the early lethality of Cdx2 null mutants. To circumvent these limitations, we used Cre-mediated conditional deletion to assess the impact of concomitant loss of Cdx1 and Cdx2 on murine primitive hematopoiesis. We found that Cdx1/Cdx2 double mutants exhibited defects in primitive hematopoiesis and yolk sac vasculature concomitant with reduced expression of several genes encoding hematopoietic transcription factors including Scl/Tal1. Chromatin immunoprecipitation analysis revealed that Scl was occupied by Cdx2 in vivo, and Cdx mutant hematopoietic yolk sac differentiation defects could be rescued by expression of exogenous Scl. These findings demonstrate critical roles for Cdx members in murine primitive hematopoiesis upstream of Scl.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fator de Transcrição CDX2/genética , Hematopoese/genética , Proteínas de Homeodomínio/genética , Neovascularização Fisiológica/genética , Proteínas Proto-Oncogênicas/biossíntese , Saco Vitelino/irrigação sanguínea , Animais , Diferenciação Celular/genética , Imunoprecipitação da Cromatina , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Knockout , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Saco Vitelino/embriologia
5.
J Biol Chem ; 292(8): 3389-3399, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28082674

RESUMO

The packaging of genomic DNA into nucleosomes creates a barrier to transcription that can be relieved through ATP-dependent chromatin remodeling via complexes such as the switch-sucrose non-fermentable (SWI-SNF) chromatin remodeling complex. The SWI-SNF complex remodels chromatin via conformational or positional changes of nucleosomes, thereby altering the access of transcriptional machinery to target genes. The SWI-SNF complex has limited ability to bind to sequence-specific elements, and, therefore, its recruitment to target loci is believed to require interaction with DNA-associated transcription factors. The Cdx family of homeodomain transcript ion factors (Cdx1, Cdx2, and Cdx4) are essential for a number of developmental programs in the mouse. Cdx1 and Cdx2 also regulate intestinal homeostasis throughout life. Although a number of Cdx target genes have been identified, the basis by which Cdx members impact their transcription is poorly understood. We have found that Cdx members interact with the SWI-SNF complex and make direct contact with Brg1, a catalytic member of SWI-SNF. Both Cdx2 and Brg1 co-occupy a number of Cdx target genes, and both factors are necessary for transcriptional regulation of such targets. Finally, Cdx2 and Brg1 occupancy occurs coincident with chromatin remodeling at some of these loci. Taken together, our findings suggest that Cdx transcription factors regulate target gene expression, in part, through recruitment of Brg1-associated SWI-SNF chromatin remodeling activity.


Assuntos
Fator de Transcrição CDX2/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Mapas de Interação de Proteínas
6.
Elife ; 52016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27823583

RESUMO

The tumor microenvironment is a critical modulator of carcinogenesis; however, in many tumor types, the influence of the stroma during preneoplastic stages is unknown. Here we explored the relationship between pre-tumor cells and their surrounding stroma in malignant progression of the cerebellar tumor medulloblastoma (MB). We show that activation of the vascular regulatory signalling axis mediated by Norrin (an atypical Wnt)/Frizzled4 (Fzd4) inhibits MB initiation in the Ptch+/- mouse model. Loss of Norrin/Fzd4-mediated signalling in endothelial cells, either genetically or by short-term blockade, increases the frequency of pre-tumor lesions and creates a tumor-permissive microenvironment at the earliest, preneoplastic stages of MB. This pro-tumor stroma, characterized by angiogenic remodelling, is associated with an accelerated transition from preneoplasia to malignancy. These data expose a stromal component that regulates the earliest stages of tumorigenesis in the cerebellum, and a novel role for the Norrin/Fzd4 axis as an endogenous anti-tumor signal in the preneoplastic niche.


Assuntos
Carcinogênese , Proteínas do Olho/metabolismo , Receptores Frizzled/metabolismo , Meduloblastoma/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Camundongos
7.
J Cell Sci ; 129(20): 3832-3844, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27632999

RESUMO

Sonic Hedgehog (Shh) is a secreted morphogen that is an essential regulator of patterning and growth. The Shh full-length protein undergoes autocleavage in the endoplasmic reticulum to generate the biologically active N-terminal fragment (ShhN), which is destined for secretion. We identified sortilin (Sort1), a member of the VPS10P-domain receptor family, as a new Shh trafficking receptor. We demonstrate that Sort-Shh interact by performing coimmunoprecipitation and proximity ligation assays in transfected cells and that they colocalize at the Golgi. Sort1 overexpression causes re-distribution of ShhN and, to a lesser extent, of full-length Shh to the Golgi and reduces Shh secretion. We show loss of Sort1 can partially rescue Hedgehog-associated patterning defects in a mouse model that is deficient in Shh processing, and we show that Sort1 levels negatively regulate anterograde Shh transport in axons in vitro and Hedgehog-dependent axon-glial interactions in vivo Taken together, we conclude that Shh and Sort1 can interact at the level of the Golgi and that Sort1 directs Shh away from the pathways that promote its secretion.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Hedgehog/metabolismo , Animais , Astrócitos/citologia , Axônios/metabolismo , Células CHO , Células COS , Proliferação de Células , Chlorocebus aethiops , Cricetinae , Cricetulus , Técnicas de Inativação de Genes , Complexo de Golgi/metabolismo , Mutação/genética , Nervo Óptico/metabolismo , Células PC12 , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transporte Proteico , Ratos , Células Ganglionares da Retina/metabolismo , Via Secretória
8.
Dev Biol ; 411(1): 85-100, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26795056

RESUMO

Neurogenesis is regulated by the dynamic and coordinated activity of several extracellular signalling pathways, but the basis for crosstalk between these pathways remains poorly understood. Here we investigated regulatory interactions between two pathways that are each required for neural progenitor cell maintenance in the postnatal retina; Hedgehog (Hh) and Notch signalling. Both pathways are activated in progenitor cells in the postnatal retina based on the co-expression of fluorescent pathway reporter transgenes at the single cell level. Disrupting Notch signalling, genetically or pharmacologically, induces a rapid downregulation of all three Gli proteins and inhibits Hh-induced proliferation. Ectopic Notch activation, while not sufficient to promote Hh signalling or proliferation, increases Gli2 protein. We show that Notch regulation of Gli2 in Müller glia renders these cells competent to proliferate in response to Hh. These data suggest that Notch signalling converges on Gli2 to prime postnatal retinal progenitor cells and Müller glia to proliferate in response to Hh.


Assuntos
Células Ependimogliais/citologia , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Receptores Notch/metabolismo , Animais , Proliferação de Células/fisiologia , Feminino , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/genética , RNA Mensageiro/genética , Receptores Notch/genética , Retina/citologia , Retina/embriologia , Transdução de Sinais , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
9.
Invest Ophthalmol Vis Sci ; 55(1): 43-54, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24194188

RESUMO

PURPOSE: The in vitro expansion of multilineage competent primary neural progenitor cells is typically limited. Hedgehog (Hh) signaling is required in vivo for the maintenance of stem cell (SC) and progenitor populations in the central nervous system, including the retina. Here we investigated the impact of Hh signaling on in vitro expansion of perinatal mouse retinal progenitor cells (RPCs). METHODS: Perinatal mouse retinal cells were treated with combinations of Hh agonist (Hh-Ag), epidermal growth factor (EGF)/fibroblast growth factor 2 (FGF2) and the cultures were assayed for long-term growth, gene expression, and dependence on Gli2. Differentiation was assessed in monolayer cultures, following in vivo transplantation and in cellular reaggregates. RESULTS: Using a combination of Hh-Ag, EGF, and FGF2, we were able to establish long-term RPC cultures (termed Hh-RPCs). The ability of this combinatorial signaling approach to block quiescence of these was not associated with altered TP53/MDM2 levels or Hh-EGF cooperativity gene expression. Efficient Hh-RPC expansion and monolayer culture establishment requires Gli2, as Hh-RPCs derived from Gli2 knockout retinal tissue fail to generate cultures that can be passaged long-term in vitro. Hedgehog RPCs retain competence for neurogenic and gliogenic differentiation in vitro; however, they fail to engraft and differentiate into retinal cell types following in vivo transplantation to the eye or in vitro when mixed with acutely dissociated perinatal retinal cells. CONCLUSIONS: Our data show that combining Hh and mitogen signaling is sufficient to promote the expansion of RPCs in vitro, but it is insufficient to maintain competence of these cells for retinal differentiation.


Assuntos
Proteínas Hedgehog/genética , Mitógenos/metabolismo , RNA/genética , Células Ganglionares da Retina/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Proteínas Hedgehog/biossíntese , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Células Ganglionares da Retina/citologia , Transdução de Sinais , Células-Tronco/citologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-25692127

RESUMO

The retina is a highly sophisticated piece of the neural machinery that begins the translation of incoming light signals into meaningful visual information. Several degenerative diseases of the retina are characterized by photoreceptor loss and eventually lead to irreversible blindness. Regenerative medicine, using tissue engineering-based constructs to deliver progenitor cells or photoreceptors along with supporting carrier matrix is a promising approach for restoration of structure and function. Fresh fibrin glue (FG) produced by the CryoSeal(®)FS system in combination with mouse retinal progenitor cells (RPCs) were evaluated in this study. In vitro expanded RPCs isolated from postnatal mouse retina were encapsulated into FG and cultured in the presence of the protease inhibitor, tranexamic acid. Encapsulation of RPCs into FG did not show adverse effects on cell proliferation or cell survival. RPCs exhibited fibroblast-like morphology concomitantly with attachment to the encapsulating FG surface. They expressed α7 and ß3 integrin subunits that could mediate attachment to fibrin matrix via an RGD-independent mechanism. The three-dimensional environment and the attachment surface provided by FG was associated with a rapid down-regulation of the progenitor marker SOX2 and enhanced the expression of the differentiation markers cone-rod homeobox and recoverin. However, the in vitro culture conditions did not promote full differentiation into mature photoreceptors. Nevertheless, we have shown that autologous fibrin, when fabricated into a scaffold for RPCs for delivery to the retina, provides the cells with external cues that could potentially improve the differentiation events. Hence, transient encapsulation of RPCs into FG could be a valid and potential treatment strategy to promote retinal regeneration following degenerative diseases. However, further optimization is necessary to maximize the outcomes in terms of mature photoreceptors.

11.
Hum Mol Genet ; 22(5): 1005-16, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23201751

RESUMO

Norrie disease (ND) is a congenital disorder characterized by retinal hypovascularization and cognitive delay. ND has been linked to mutations in 'Norrie Disease Protein' (Ndp), which encodes the secreted protein Norrin. Norrin functions as a secreted angiogenic factor, although its role in neural development has not been assessed. Here, we show that Ndp expression is initiated in retinal progenitors in response to Hedgehog (Hh) signaling, which induces Gli2 binding to the Ndp promoter. Using a combination of genetic epistasis and acute RNAi-knockdown approaches, we show that Ndp is required downstream of Hh activation to induce retinal progenitor proliferation in the retina. Strikingly, Ndp regulates the rate of cell-cycle re-entry and not cell-cycle kinetics, thereby uncoupling the self-renewal and cell-cycle progression functions of Hh. Taken together, we have uncovered a cell autonomous function for Ndp in retinal progenitor proliferation that is independent of its function in the retinal vasculature, which could explain the neural defects associated with ND.


Assuntos
Cegueira/congênito , Proteínas do Olho/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas do Tecido Nervoso/genética , Doenças do Sistema Nervoso/genética , Retina/metabolismo , Espasmos Infantis/genética , Via de Sinalização Wnt , Animais , Cegueira/genética , Cegueira/metabolismo , Ciclo Celular/genética , Proliferação de Células , Epistasia Genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Doenças Genéticas Ligadas ao Cromossomo X , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/metabolismo , Ligação Proteica , Retina/crescimento & desenvolvimento , Degeneração Retiniana , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/metabolismo , Espasmos Infantis/metabolismo , Via de Sinalização Wnt/genética , Proteína Gli2 com Dedos de Zinco
12.
J Neurosci ; 31(13): 5169-80, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21451052

RESUMO

The morphogen sonic hedgehog (Shh) plays a crucial role in development of the CNS, including the neural retina. Suppressor of fused (Sufu) has been recently identified as a critical regulator of Hh signaling in mammals. However, the precise roles that Sufu plays in the regulation of proliferation and cell-fate decisions in neural progenitors is unknown. Here, we have addressed these questions by conditionally deleting Sufu in mouse multipotent retinal progenitor cells (RPCs). Sufu deletion in RPCs results in transient increases in Hh activity and proliferation followed by developmentally premature cell-cycle exit. Importantly, we demonstrate a novel role for Sufu in the maintenance of multipotency in RPCs. Sufu-null RPCs downregulate transcription factors required to specify or maintain RPC identity (Rax, Vsx2) and multipotency (Pax6) but continue to express the neural progenitor marker Sox2. These cells fail to express retinal lineage-specific transcription factors, such as Math5, and adopt an amacrine or horizontal cell fate at the expense of all other classes of retinal neurons. Genetic elimination of Gli2 in Sufu-null RPCs attenuates Hh pathway activity and restores multipotency in neural progenitors. These data provide novel evidence that Sufu-mediated antagonism of Hh/Gli2 signaling is required to maintain RPC multipotency and identity.


Assuntos
Células-Tronco Multipotentes/fisiologia , Neurônios/fisiologia , Proteínas Repressoras/fisiologia , Retina/embriologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Regulação para Baixo/genética , Feminino , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Gravidez , Distribuição Aleatória , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Retina/citologia , Retina/fisiologia , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...