Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33999993

RESUMO

In mammals, the pre-gastrula proximal epiblast gives rise to primordial germ cells (PGCs) or somatic precursors in response to BMP4 and WNT signaling. Entry into the germline requires activation of a naïve-like pluripotency gene regulatory network (GRN). Recent work has shown that suppression of OTX2 expression in the epiblast by BMP4 allows cells to develop a PGC fate in a precise temporal window. However, the mechanisms by which OTX2 suppresses PGC fate are unknown. Here, we show that, in mice, OTX2 prevents epiblast cells from activating the pluripotency GRN by direct repression of Oct4 and Nanog. Loss of this control during PGC differentiation in vitro causes widespread activation of the pluripotency GRN and a deregulated response to LIF, BMP4 and WNT signaling. These abnormalities, in specific cell culture conditions, result in massive germline entry at the expense of somatic mesoderm differentiation. Increased generation of PGCs also occurs in mutant embryos. We propose that the OTX2-mediated repressive control of Oct4 and Nanog is the basis of the mechanism that determines epiblast contribution to germline and somatic lineage.


Assuntos
Células Germinativas/citologia , Camadas Germinativas/citologia , Proteína Homeobox Nanog/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fatores de Transcrição Otx/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Pluripotentes/citologia , Via de Sinalização Wnt/fisiologia
2.
Cell Rep ; 23(12): 3635-3646, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29925004

RESUMO

Placental growth factor (PlGF) is a proangiogenic member of the vascular endothelial growth factor (VEGF) family playing a central role in pathological angiogenesis. PlGF-DE is a PlGF variant unable to bind vascular endothelial growth factor receptor 1 (VEGFR-1) but still able to generate heterodimer with VEGF-A. We have generated PlGF-DE knockin mice that are vital and fertile and show unaltered expression of Plgf, Vegf-a, Vegfr-1, and Vegfr-2 compared with wild-type mice. Interestingly, these mutants showed additional and remarkable angiogenesis impairment in tumor growth, hindlimb ischemia, and choroidal neovascularization compared with both PlGF knockout and wild-type mice. These findings provided insights on VEGF-A/PlGF heterodimer function, which was able to rescue neovascularization and vascular leakage in PlGF-DE knockin mice. Collectively, these data show that PlGF-DE knockin mouse could be considered the full functional knockout of PlGF, suggesting a reassessment of the phenotypes of knockout mice for the genes whose products are able to generate heterodimeric proteins.


Assuntos
Técnicas de Introdução de Genes , Fator de Crescimento Placentário/metabolismo , Multimerização Proteica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Stem Cell Reports ; 9(5): 1642-1659, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29056334

RESUMO

Embryonic stem cells (ESCs) cultured in leukemia inhibitory factor (LIF) plus fetal bovine serum (FBS) exhibit heterogeneity in the expression of naive and primed transcription factors. This heterogeneity reflects the dynamic condition of ESCs and their versatility to promptly respond to signaling effectors promoting naive or primed pluripotency. Here, we report that ESCs lacking Nanog or overexpressing Otx2 exhibit an early primed identity in LIF + FBS and fail to convert into 2i-induced naive state. Conversely, Otx2-null ESCs possess naive identity features in LIF + FBS similar to Nanog-overexpressing ESCs and convert poorly into FGF-induced early primed state. When both Nanog and Otx2 are inactivated, ESCs cultured in LIF + FBS exhibit primed identity and weakened ability to convert into naive state. These data suggest that, through mutual antagonism, NANOG and OTX2 specify the heterogeneous identity of ESCs cultured in LIF + FBS and individually predispose them for optimal response to naive or primed inducing factors.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Murinas/citologia , Proteína Homeobox Nanog/genética , Fatores de Transcrição Otx/genética , Animais , Linhagem Celular , Meios de Cultura Livres de Soro/farmacologia , Fator Inibidor de Leucemia/farmacologia , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Proteína Homeobox Nanog/metabolismo , Fatores de Transcrição Otx/metabolismo
4.
Nat Commun ; 7: 12589, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27586544

RESUMO

Known molecular determinants of developmental plasticity are mainly transcription factors, while the extrinsic regulation of this process has been largely unexplored. Here we identify Cripto as one of the earliest epiblast markers and a key extracellular determinant of the naive and primed pluripotent states. We demonstrate that Cripto sustains mouse embryonic stem cell (ESC) self-renewal by modulating Wnt/ß-catenin, whereas it maintains mouse epiblast stem cell (EpiSC) and human ESC pluripotency through Nodal/Smad2. Moreover, we provide unprecedented evidence that Cripto controls the metabolic reprogramming in ESCs to EpiSC transition. Remarkably, Cripto deficiency attenuates ESC lineage restriction in vitro and in vivo, and permits ESC transdifferentiation into trophectoderm lineage, suggesting that Cripto has earlier functions than previously recognized. All together, our studies provide novel insights into the current model of mammalian pluripotency and contribute to the understanding of the extrinsic regulation of the first cell lineage decision in the embryo.


Assuntos
Desenvolvimento Embrionário/fisiologia , Células-Tronco Embrionárias/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Pluripotentes/fisiologia , beta Catenina/metabolismo , Animais , Reprogramação Celular/genética , Fator de Crescimento Epidérmico/genética , Camadas Germinativas/citologia , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteína Nodal/metabolismo , Proteína Smad2/metabolismo , Proteínas Wnt/metabolismo
5.
Cell Rep ; 15(12): 2651-64, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27292645

RESUMO

Mouse embryonic stem cells (ESCs) and the inner cell mass (ICM)-derived epiblast exhibit naive pluripotency. ESC-derived epiblast stem cells (EpiSCs) and the postimplantation epiblast exhibit primed pluripotency. Although core pluripotency factors are well-characterized, additional regulators, including Otx2, recently have been shown to function during the transition from naive to primed pluripotency. Here we uncover a role for Otx2 in the control of the naive pluripotent state. We analyzed Otx2-binding activity in ESCs and EpiSCs and identified Nanog, Oct4, and Sox2 as direct targets. To unravel the Otx2 transcriptional network, we targeted the strongest Otx2-binding site in the Nanog promoter, finding that this site modulates the size of specific ESC-subtype compartments in cultured cells and promotes Nanog expression in vivo, predisposing ICM differentiation to epiblast. Otx2-mediated Nanog regulation thus contributes to the integrity of the ESC state and cell lineage specification in preimplantation development.


Assuntos
Blastocisto/citologia , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Proteína Homeobox Nanog/genética , Fatores de Transcrição Otx/metabolismo , Regiões Promotoras Genéticas/genética , Animais , Sítios de Ligação , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Compartimento Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimera/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camadas Germinativas/efeitos dos fármacos , Camadas Germinativas/metabolismo , Fator Inibidor de Leucemia/farmacologia , Mesoderma/citologia , Camundongos , Mutação/genética , Proteína Homeobox Nanog/metabolismo , Fatores de Transcrição Otx/genética , Ligação Proteica/efeitos dos fármacos
6.
EMBO Mol Med ; 4(3): 192-205, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22247000

RESUMO

Ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, which is characterized by cleft palate and severe defects of the skin, is an autosomal dominant disorder caused by mutations in the gene encoding transcription factor p63. Here, we report the generation of a knock-in mouse model for AEC syndrome (p63(+/L514F) ) that recapitulates the human disorder. The AEC mutation exerts a selective dominant-negative function on wild-type p63 by affecting progenitor cell expansion during ectodermal development leading to a defective epidermal stem cell compartment. These phenotypes are associated with impairment of fibroblast growth factor (FGF) signalling resulting from reduced expression of Fgfr2 and Fgfr3, direct p63 target genes. In parallel, a defective stem cell compartment is observed in humans affected by AEC syndrome and in Fgfr2b(-/-) mice. Restoring Fgfr2b expression in p63(+/L514F) epithelial cells by treatment with FGF7 reactivates downstream mitogen-activated protein kinase signalling and cell proliferation. These findings establish a functional link between FGF signalling and p63 in the expansion of epithelial progenitor cells and provide mechanistic insights into the pathogenesis of AEC syndrome.


Assuntos
Proliferação de Células , Fenda Labial/metabolismo , Fissura Palatina/metabolismo , Ectoderma/citologia , Displasia Ectodérmica/metabolismo , Anormalidades do Olho/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Animais , Fenda Labial/genética , Fenda Labial/fisiopatologia , Fissura Palatina/genética , Fissura Palatina/fisiopatologia , Ectoderma/metabolismo , Displasia Ectodérmica/genética , Displasia Ectodérmica/fisiopatologia , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Pálpebras/anormalidades , Pálpebras/metabolismo , Pálpebras/fisiopatologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
7.
Am J Pathol ; 175(6): 2609-17, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19893048

RESUMO

The roles in brain development. Previous studies have shown the association between OTX2 and OTX1 with anaplastic and desmoplastic medulloblastomas, respectively. Here, we investigated OTX1 and OTX2 expression in Non-Hodgkin Lymphoma (NHL) and multiple myeloma. A combination of semiquantitative RT-PCR, Western blot, and immunohistochemical analyses was used to measure OTX1 and OTX2 levels in normal lymphoid tissues and in 184 tumor specimens representative of various forms of NHL and multiple myeloma. OTX1 expression was activated in 94% of diffuse large B-cell lymphomas, in all Burkitt lymphomas, and in 90% of high-grade follicular lymphomas. OTX1 was undetectable in precursor-B lymphoblastic lymphoma, chronic lymphocytic leukemia, and in most marginal zone and mantle cell lymphomas and multiple myeloma. OTX2 was undetectable in all analyzed malignancies. Analysis of OTX1 expression in normal lymphoid tissues identified a subset of resting germinal center (GC) B cells lacking PAX5 and BCL6 and expressing cytoplasmic IgG and syndecan. About 50% of OTX1(+) GC B cells co-expressed CD10 and CD20. This study identifies OTX1 as a molecular marker for high-grade GC-derived NHL and suggests an involvement of this transcription factor in B-cell lymphomagenesis. Furthermore, OTX1 expression in a subset of normal GC B cells carrying plasma cell markers suggests its possible contribution to terminal B-cell differentiation.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Biomarcadores Tumorais/análise , Centro Germinativo/metabolismo , Linfoma não Hodgkin/metabolismo , Fatores de Transcrição Otx/biossíntese , Western Blotting , Humanos , Imuno-Histoquímica , Mieloma Múltiplo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Development ; 135(20): 3459-70, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18820178

RESUMO

Meso-diencephalic dopaminergic (mdDA) neurons control voluntary movement, cognition and the reward response, and their degeneration is associated with Parkinson's disease (PD). Prospective cell transplantation therapies for PD require full knowledge of the developmental pathways that control mdDA neurogenesis. We have previously shown that Otx2 is required for the establishment of the mesencephalic field and molecular code of the entire ventral mesencephalon (VM). Here, we investigate whether Otx2 is a specific determinant of mesencephalic dopaminergic (mesDA) neurogenesis by studying mouse mutants that conditionally overexpress or lack Otx2. Our data show that Otx2 overexpression in the VM causes a dose-dependent and selective increase in both mesDA progenitors and neurons, which correlates with a remarkable and specific enhancement in the proliferating activity of mesDA progenitors. Consistently, lack of Otx2 in the VM specifically affects the proliferation of Sox2+ mesDA progenitors and causes their premature post-mitotic transition. Analysis of the developmental pathway that controls the differentiation of mesDA neurons shows that, in the absence of Otx2, the expression of Lmx1a and Msx1, and the proneural genes Ngn2 and Mash1 is not activated in Sox2+ mesDA progenitors, which largely fail to differentiate into Nurr1+ mesDA precursors. Furthermore, proliferation and differentiation abnormalities exhibit increasing severity along the anterior-posterior (AP) axis of the VM. These findings demonstrate that Otx2, through an AP graded effect, is intrinsically required to control proliferation and differentiation of mesDA progenitors. Thus, our data provide new insights into the mechanism of mesDA neuron specification and suggest Otx2 as a potential target for cell replacement-based therapeutic approaches in PD.


Assuntos
Dopamina/metabolismo , Células-Tronco Embrionárias/citologia , Mesencéfalo/citologia , Mesencéfalo/embriologia , Fatores de Transcrição Otx/fisiologia , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Embrião de Mamíferos , Imuno-Histoquímica , Hibridização In Situ , Mesencéfalo/metabolismo , Camundongos , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo
9.
Mol Cell Neurosci ; 31(2): 293-302, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16256364

RESUMO

Here, we have investigated the neurological consequences of restricted inactivation of Otx2 in adult En1(cre/+); Otx2(flox/flox) mice. In agreement with the crucial role of Otx2 in midbrain patterning, the mutants had a substantial reduction in tyrosine hydroxylase containing neurons. Although the reduction in the number of DAergic neurons was comparable between the SNc and the VTA, we found an unexpected selectivity in the deinnervation of the terminal fields affecting preferentially the ventral striatum and the olfactory tubercle. Interestingly, the mutants showed no abnormalities in exploratory activity or motor coordination. However, the absence of normal DA tone generated significant alterations in DA D1-receptor signalling as indicated by increased mutant striatal levels of phosphorylated DARPP-32 and by an altered motor response to amphetamine. Therefore, we suggest that the En1(cre/+); Otx2(flox/flox) mutant mouse model represents a genetic tool for investigating molecular and behavioural consequences of developmental neuronal dysfunction in the DAergic system.


Assuntos
Anfetaminas/metabolismo , Dopamina/metabolismo , Mesencéfalo/embriologia , Morfogênese , Fatores de Transcrição Otx , Anfetaminas/farmacologia , Animais , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Feminino , Inativação Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutação , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Receptores de Dopamina D1/metabolismo , Teste de Desempenho do Rota-Rod , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Proc Natl Acad Sci U S A ; 100(18): 10299-303, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12934017

RESUMO

Otx1 belongs to the paired class of homeobox genes and plays a pivotal role in brain development. Here, we show that Otx1 is expressed in hematopoietic pluripotent and erythroid progenitor cells. Moreover, bone marrow cells from mice lacking Otx1 exhibit a cell-autonomous impairment of the erythroid compartment. In agreement with these results, molecular analysis revealed decreased levels of erythroid genes that include the SCL and GATA-1 transcription factors. Accordingly, a gain of function of SCL rescues the erythroid deficiency in Otx1-/- mice. Taken together, our findings indicate a function for Otx1 in the regulation of blood cell production.


Assuntos
Hematopoese , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Ligação a DNA/genética , Eritropoese , Feminino , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fatores de Transcrição Otx , Proteínas Proto-Oncogênicas/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA