Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Open Biol ; 14(4): 230383, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38629124

RESUMO

Non-clustered protocadherins (ncPcdhs) are adhesive molecules with spatio-temporally regulated overlapping expression in the developing nervous system. Although their unique role in neurogenesis has been widely studied, their combinatorial role in brain physiology and pathology is poorly understood. Using probabilistic cell typing by in situ sequencing, we demonstrate combinatorial inter- and intra-familial expression of ncPcdhs in the developing mouse cortex and hippocampus, at single-cell resolution. We discovered the combinatorial expression of Protocadherin-19 (Pcdh19), a protein involved in PCDH19-clustering epilepsy, with Pcdh1, Pcdh9 or Cadherin 13 (Cdh13) in excitatory neurons. Using aggregation assays, we demonstrate a code-specific adhesion function of PCDH19; mosaic PCDH19 absence in PCDH19+9 and PCDH19 + CDH13, but not in PCDH19+1 codes, alters cell-cell interaction. Interestingly, we found that PCDH19 as a dominant protein in two heterophilic adhesion codes could promote trans-interaction between them. In addition, we discovered increased CDH13-mediated cell adhesion in the presence of PCDH19, suggesting a potential role of PCDH19 as an adhesion mediator of CDH13. Finally, we demonstrated novel cis-interactions between PCDH19 and PCDH1, PCDH9 and CDH13. These observations suggest that there is a unique combinatorial code with a cell- and region-specific characteristic where a single molecule defines the heterophilic cell-cell adhesion properties of each code.


Assuntos
Epilepsia , Protocaderinas , Camundongos , Animais , Adesão Celular , Encéfalo , Epilepsia/metabolismo , Neurônios/metabolismo
2.
iScience ; 25(11): 105317, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36310582

RESUMO

Immunotherapy for cancer that aims to promote T cell anti-tumor activity has changed current clinical practice, where some previously lethal cancers have now become treatable. However, clinical trials with low response rates have been disappointing for pancreatic ductal adenocarcinoma (PDAC). One suggested explanation is the accumulation of dominantly immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells in the tumor microenvironment (TME). Using retrospectively collected tumor specimens and transcriptomic data from PDAC, we demonstrate that expression of the scavenger receptor MARCO correlates with poor prognosis and a lymphocyte-excluding tumor phenotype. PDAC cell lines produce IL-10 and induce high expression of MARCO in myeloid cells, and this was further enhanced during hypoxic conditions. These myeloid cells suppressed effector T and natural killer (NK) cells and blocked NK cell tumor infiltration and tumor killing in a PDAC 3D-spheroid model. Anti-human MARCO (anti-hMARCO) antibody targeting triggered the repolarization of tumor-associated macrophages and activated the inflammasome machinery, resulting in IL-18 production. This in turn enhanced T cell and NK cell functions. The targeting of MARCO thus remodels the TME and represents a rational approach to make immunotherapy more efficient in PDAC patients.

3.
Development ; 149(14)2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35861233

RESUMO

The transcription factor SOX2 is a vital regulator of stem cell activity in various developing and adult tissues. Mounting evidence has demonstrated the importance of SOX2 in regulating the induction and maintenance of stemness as well as in controlling cell proliferation, lineage decisions and differentiation. Recent studies have revealed that the ability of SOX2 to regulate these stem cell features involves its function as a pioneer factor, with the capacity to target nucleosomal DNA, modulate chromatin accessibility and prepare silent genes for subsequent activation. Moreover, although SOX2 binds to similar DNA motifs in different stem cells, its multifaceted and cell type-specific functions are reliant on context-dependent features. These cell type-specific properties include variations in partner factor availability and SOX2 protein expression levels. In this Primer, we discuss recent findings that have increased our understanding of how SOX2 executes its versatile functions as a master regulator of stem cell activities.


Assuntos
Nucleossomos , Fatores de Transcrição SOXB1 , Diferenciação Celular/genética , Proliferação de Células/genética , Cromatina , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
4.
Bioessays ; 43(7): e2000285, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34008221

RESUMO

Development and maintenance of diverse organ systems require context-specific regulation of stem cell behaviour. We hypothesize that this is achieved via reciprocal regulation between the cell cycle machinery and differentiation factors. This idea is supported by the parallel evolutionary emergence of differentiation pathways, cell cycle components and complex multicellularity. In addition, the activities of different cell cycle phases have been found to bias cells towards stem cell maintenance or differentiation. Finally, several direct mechanistic links between these two processes have been established. Here, we focus on interactions between cyclin-CDK complexes and differentiation regulators of the Notch pathway and Sox family of transcription factors within the context of pluripotent and neural stem cells. Thus, this hypothesis formalizes the links between these two processes as an integrated network. Since such factors are common to all stem cells, better understanding their interconnections will help to explain their behaviour in health and disease.


Assuntos
Ciclinas , Células-Tronco Neurais , Ciclo Celular , Diferenciação Celular , Quinases Ciclina-Dependentes
5.
Nat Commun ; 11(1): 2898, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32518258

RESUMO

The sequential generation of layer-specific cortical neurons requires radial glia cells (RGCs) to precisely balance self-renewal and lineage commitment. While specific cell-cycle phases have been associated with these decisions, the mechanisms linking the cell-cycle machinery to cell-fate commitment remain obscure. Using single-cell RNA-sequencing, we find that the strongest transcriptional signature defining multipotent RGCs is that of G2/M-phase, and particularly CYCLIN-B1/2, while lineage-committed progenitors are enriched in G1/S-phase genes, including CYCLIN-D1. These data also reveal cell-surface markers that allow us to isolate RGCs and lineage-committed progenitors, and functionally confirm the relationship between cell-cycle phase enrichment and cell fate competence. Finally, we use cortical electroporation to demonstrate that CYCLIN-B1/2 cooperate with CDK1 to maintain uncommitted RGCs by activating the NOTCH pathway, and that CYCLIN-D1 promotes differentiation. Thus, this work establishes that cell-cycle phase-specific regulators act in opposition to coordinate the self-renewal and lineage commitment of RGCs via core stem cell regulatory pathways.


Assuntos
Ciclina B1/fisiologia , Ciclina B2/fisiologia , Ciclina D1/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Proteína Quinase CDC2/fisiologia , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Separação Celular , Córtex Cerebral/embriologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Células-Tronco/citologia
6.
Proc Natl Acad Sci U S A ; 116(34): 16997-17006, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31375625

RESUMO

Despite the discovery of the oxygen-sensitive regulation of HIFα by the von Hippel-Lindau (VHL) protein, the mechanisms underlying the complex genotype/phenotype correlations in VHL disease remain unknown. Some germline VHL mutations cause familial pheochromocytoma and encode proteins that preserve their ability to down-regulate HIFα. While type 1, 2A, and 2B VHL mutants are defective in regulating HIFα, type 2C mutants encode proteins that preserve their ability to down-regulate HIFα. Here, we identified an oxygen-sensitive function of VHL that is abolished by VHL type 2C mutations. We found that BIM-EL, a proapoptotic BH3-only protein, is hydroxylated by EglN3 and subsequently bound by VHL. VHL mutants fail to bind hydroxylated BIM-EL, regardless of whether they have the ability to bind hydroxylated HIFα or not. VHL binding inhibits BIM-EL phosphorylation by extracellular signal-related kinase (ERK) on serine 69. This causes BIM-EL to escape from proteasomal degradation, allowing it to enhance EglN3-induced apoptosis. BIM-EL was rapidly degraded in cells lacking wild-type VHL or in which EglN3 was inactivated genetically or by lack of oxygen, leading to enhanced cell survival and chemotherapy resistance. Combination therapy using ERK inhibitors, however, resensitizes VHL- and EglN3-deficient cells that are otherwise cisplatin-resistant.


Assuntos
Neoplasias das Glândulas Suprarrenais , Proteína 11 Semelhante a Bcl-2/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mutação , Feocromocitoma , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína 11 Semelhante a Bcl-2/genética , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Hidroxilação/efeitos dos fármacos , Hidroxilação/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Células PC12 , Feocromocitoma/tratamento farmacológico , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Proteólise/efeitos dos fármacos , Ratos , Proteína Supressora de Tumor Von Hippel-Lindau/genética
7.
EMBO Rep ; 19(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30166336

RESUMO

SOX transcription factors have important roles during astrocyte and oligodendrocyte development, but how glial genes are specified and activated in a sub-lineage-specific fashion remains unknown. Here, we define glial-specific gene expression in the developing spinal cord using single-cell RNA-sequencing. Moreover, by ChIP-seq analyses we show that these glial gene sets are extensively preselected already in multipotent neural precursor cells through prebinding by SOX3. In the subsequent lineage-restricted glial precursor cells, astrocyte genes become additionally targeted by SOX9 at DNA regions strongly enriched for Nfi binding motifs. Oligodendrocyte genes instead are prebound by SOX9 only, at sites which during oligodendrocyte maturation are targeted by SOX10. Interestingly, reporter gene assays and functional studies in the spinal cord reveal that SOX3 binding represses the synergistic activation of astrocyte genes by SOX9 and NFIA, whereas oligodendrocyte genes are activated in a combinatorial manner by SOX9 and SOX10. These genome-wide studies demonstrate how sequentially expressed SOX proteins act on lineage-specific regulatory DNA elements to coordinate glial gene expression both in a temporal and in a sub-lineage-specific fashion.


Assuntos
Astrócitos/fisiologia , Oligodendroglia/fisiologia , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOXB1/genética , Medula Espinal/citologia , Animais , Diferenciação Celular/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Neurais , Neuroglia/citologia , Neuroglia/fisiologia , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Medula Espinal/crescimento & desenvolvimento
8.
PLoS Genet ; 14(2): e1007224, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29432416

RESUMO

Stem cells are defined by their capacities to self-renew and generate progeny of multiple lineages. The transcription factor SOX2 has key roles in the regulation of stem cell characteristics, but whether SOX2 achieves these functions through similar mechanisms in distinct stem cell populations is not known. To address this question, we performed RNA-seq and SOX2 ChIP-seq on embryonic mouse cortex, spinal cord, stomach and lung/esophagus. We demonstrate that, although SOX2 binds a similar motif in the different cell types, its target regions are primarily cell-type-specific and enriched for the distinct binding motifs of appropriately expressed interacting co-factors. Furthermore, cell-type-specific SOX2 binding in endodermal and neural cells is most often found around genes specifically expressed in the corresponding tissue. Consistent with this, we demonstrate that SOX2 target regions can act as cis-regulatory modules capable of directing reporter expression to appropriate tissues in a zebrafish reporter assay. In contrast, SOX2 binding sites found in both endodermal and neural tissues are associated with genes regulating general stem cell features, such as proliferation. Notably, we provide evidence that SOX2 regulates proliferation through conserved mechanisms and target genes in both germ layers examined. Together, these findings demonstrate how SOX2 simultaneously regulates cell-type-specific, as well as core transcriptional programs in neural and endodermal stem cells.


Assuntos
Sistema Nervoso Central/embriologia , Endoderma/citologia , Endoderma/embriologia , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Neurais/fisiologia , Organogênese/genética , Fatores de Transcrição SOXB1/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Sistema Nervoso Central/citologia , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição SOXB1/genética
9.
Cancer Res ; 77(18): 4985-4997, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687615

RESUMO

Molecular mechanisms preventing self-renewing brain stem cells from oncogenic transformation are poorly defined. We show that the expression levels of SOX5, SOX6, and SOX21 (SOX5/6/21) transcription factors increase in stem cells of the subventricular zone (SVZ) upon oncogenic stress, whereas their expression in human glioma decreases during malignant progression. Elevated levels of SOX5/6/21 promoted SVZ cells to exit the cell cycle, whereas genetic ablation of SOX5/6/21 dramatically increased the capacity of these cells to form glioma-like tumors in an oncogene-driven mouse brain tumor model. Loss-of-function experiments revealed that SOX5/6/21 prevent detrimental hyperproliferation of oncogene expressing SVZ cells by facilitating an antiproliferative expression profile. Consistently, restoring high levels of SOX5/6/21 in human primary glioblastoma cells enabled expression of CDK inhibitors and decreased p53 protein turnover, which blocked their tumorigenic capacity through cellular senescence and apoptosis. Altogether, these results provide evidence that SOX5/6/21 play a central role in driving a tumor suppressor response in brain stem cells upon oncogenic insult. Cancer Res; 77(18); 4985-97. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/patologia , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB2/metabolismo , Fatores de Transcrição SOXD/fisiologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Senescência Celular , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fatores de Transcrição SOXB2/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cell Rep ; 17(2): 469-483, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27705795

RESUMO

The generation of neurons from neural stem cells requires large-scale changes in gene expression that are controlled to a large extent by proneural transcription factors, such as Ascl1. While recent studies have characterized the differentiation genes activated by proneural factors, less is known on the mechanisms that suppress progenitor cell identity. Here, we show that Ascl1 induces the transcription factor MyT1 while promoting neuronal differentiation. We combined functional studies of MyT1 during neurogenesis with the characterization of its transcriptional program. MyT1 binding is associated with repression of gene transcription in neural progenitor cells. It promotes neuronal differentiation by counteracting the inhibitory activity of Notch signaling at multiple levels, targeting the Notch1 receptor and many of its downstream targets. These include regulators of the neural progenitor program, such as Hes1, Sox2, Id3, and Olig1. Thus, Ascl1 suppresses Notch signaling cell-autonomously via MyT1, coupling neuronal differentiation with repression of the progenitor fate.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Neurogênese/genética , Receptor Notch1/genética , Fatores de Transcrição/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Inibidoras de Diferenciação/genética , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fatores de Transcrição SOXB1/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição HES-1/genética , Vertebrados/genética , Vertebrados/crescimento & desenvolvimento
11.
Genome Res ; 26(7): 908-17, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27197220

RESUMO

Spatially distinct gene expression profiles in neural stem cells (NSCs) are a prerequisite to the formation of neuronal diversity, but how these arise from the regulatory interactions between chromatin accessibility and transcription factor activity has remained unclear. Here, we demonstrate that, despite their distinct gene expression profiles, NSCs of the mouse cortex and spinal cord share the majority of their DNase I hypersensitive sites (DHSs). Regardless of this similarity, domain-specific gene expression is highly correlated with the relative accessibility of associated DHSs, as determined by sequence read density. Notably, the binding pattern of the general NSC transcription factor SOX2 is also largely cell type specific and coincides with an enrichment of LHX2 motifs in the cortex and HOXA9 motifs in the spinal cord. Interestingly, in a zebrafish reporter gene system, these motifs were critical determinants of patterned gene expression along the rostral-caudal axis. Our findings establish a predictive model for patterned NSC gene expression, whereby domain-specific expression of LHX2 and HOX proteins act on their target motifs within commonly accessible cis-regulatory regions to specify SOX2 binding. In turn, this binding correlates strongly with these DHSs relative accessibility-a robust predictor of neighboring gene expression.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Neurais/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo , Medula Espinal/citologia , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
12.
Cell Rep ; 9(5): 1908-1920, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25482558

RESUMO

Organ formation and maintenance depends on slowly self-renewing stem cells that supply an intermediate population of rapidly dividing progenitors, but how this proliferative hierarchy is regulated is unknown. By performing genome-wide single-cell and functional analyses in the cortex, we demonstrate that reduced Sox2 expression is a key regulatory signature of the transition between stem cells and rapidly dividing progenitors. In stem cells, Sox2 is expressed at high levels, which enables its repression of proproliferative genes, of which Cyclin D1 is the most potent target. Sox2 confers this function through binding to low-affinity motifs, which facilitate the recruitment of Gro/Tle corepressors in synergy with Tcf/Lef proteins. Upon differentiation, proneural factors reduce Sox2 expression, which derepresses Cyclin D1 and promotes proliferation. Our results show how concentration-dependent Sox2 occupancy of DNA motifs of varying affinities translates into recruitment of repressive complexes, which regulate the proliferative dynamics of neural stem and progenitor cells.


Assuntos
Proliferação de Células , Córtex Cerebral/citologia , Células-Tronco Neurais/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Ciclo Celular , Diferenciação Celular , Ciclina D1/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo
13.
Cell Rep ; 8(4): 1018-25, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25127144

RESUMO

Distinct midbrain dopamine (mDA) neuron subtypes are found in the substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA), but it is mainly SNc neurons that degenerate in Parkinson's disease. Interest in how mDA neurons develop has been stimulated by the potential use of stem cells in therapy or disease modeling. However, very little is known about how specific dopaminergic subtypes are generated. Here, we show that the expression profiles of the transcription factors Sox6, Otx2, and Nolz1 define subpopulations of mDA neurons already at the neural progenitor cell stage. After cell-cycle exit, Sox6 selectively localizes to SNc neurons, while Otx2 and Nolz1 are expressed in a subset of VTA neurons. Importantly, Sox6 ablation leads to decreased expression of SNc markers and a corresponding increase in VTA markers, while Otx2 ablation has the opposite effect. Moreover, deletion of Sox6 affects striatal innervation and dopamine levels. We also find reduced Sox6 levels in Parkinson's disease patients. These findings identify Sox6 as a determinant of SNc neuron development and should facilitate the engineering of relevant mDA neurons for cell therapy and disease modeling.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Fatores de Transcrição Otx/fisiologia , Fatores de Transcrição SOXD/fisiologia , Substância Negra/citologia , Área Tegmentar Ventral/citologia , Animais , Padronização Corporal , Humanos , Camundongos Transgênicos , Especificidade de Órgãos , Substância Negra/embriologia , Substância Negra/metabolismo , Área Tegmentar Ventral/embriologia , Área Tegmentar Ventral/metabolismo
14.
Proc Natl Acad Sci U S A ; 111(7): 2794-9, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24501124

RESUMO

How a pool of undifferentiated neural progenitor cells is maintained in the developing nervous system is an issue that remains unresolved. One of the key transcription factors for self-renewal of these cells is Sox2, the forced expression of which has been shown to inhibit neuronal differentiation in vivo. To dissect the molecular mechanisms of Sox2 activity, a ChIP-on-chip assay has been carried out for Sox2, and multiple candidate direct target genes have been isolated. In this report, we provide evidence indicating that Sox6, which like Sox2 belongs to the SRY-related HMG box transcription factor family, is a bona-fide direct regulatory target of Sox2. In vivo, Sox6 expression is seen with a temporal lag in Sox2-positive neural precursor cells in the ventricular zone, and Sox2 promotes expression of Sox6 as a transcriptional activator. Interestingly, gain- and loss-of-function assays indicate that Sox6 in turn is required for the maintenance of Sox2 expression, suggesting that a positive feedback loop, which functions to inhibit premature neuronal differentiation, exists between the two transcription factors.


Assuntos
Diferenciação Celular/fisiologia , Sistema Nervoso Central/embriologia , Retroalimentação Fisiológica/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXD/metabolismo , Animais , Sequência de Bases , Embrião de Galinha , Imunoprecipitação da Cromatina , Primers do DNA/genética , Imuno-Histoquímica , Hibridização In Situ , Dados de Sequência Molecular , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXD/genética , Análise de Sequência de DNA
15.
Proc Natl Acad Sci U S A ; 110(18): 7330-5, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23589857

RESUMO

The reiterative deployment of a small cadre of morphogen signals underlies patterning and growth of most tissues during embyogenesis, but how such inductive events result in tissue-specific responses remains poorly understood. By characterizing cis-regulatory modules (CRMs) associated with genes regulated by Sonic hedgehog (Shh), retinoids, or bone morphogenetic proteins in the CNS, we provide evidence that the neural-specific interpretation of morphogen signaling reflects a direct integration of these pathways with SoxB1 proteins at the CRM level. Moreover, expression of SoxB1 proteins in the limb bud confers on mesodermal cells the potential to activate neural-specific target genes upon Shh, retinoid, or bone morphogenetic protein signaling, and the collocation of binding sites for SoxB1 and morphogen-mediatory transcription factors in CRMs faithfully predicts neural-specific gene activity. Thus, an unexpectedly simple transcriptional paradigm appears to conceptually explain the neural-specific interpretation of pleiotropic signaling during vertebrate development. Importantly, genes induced in a SoxB1-dependent manner appear to constitute repressive gene regulatory networks that are directly interlinked at the CRM level to constrain the regional expression of patterning genes. Accordingly, not only does the topology of SoxB1-driven gene regulatory networks provide a tissue-specific mode of gene activation, but it also determines the spatial expression pattern of target genes within the developing neural tube.


Assuntos
Redes Reguladoras de Genes/genética , Neurônios/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/genética , Animais , Sítios de Ligação , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Galinhas , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Genoma/genética , Proteínas Hedgehog/metabolismo , Botões de Extremidades/efeitos dos fármacos , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Camundongos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Sequências Reguladoras de Ácido Nucleico/genética , Retinoides/farmacologia , Transdução de Sinais/efeitos dos fármacos
16.
Dev Cell ; 23(5): 1006-19, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23153497

RESUMO

Morphogens orchestrate tissue patterning in a concentration-dependent fashion during vertebrate embryogenesis, yet little is known of how positional information provided by such signals is translated into discrete transcriptional outputs. Here we have identified and characterized cis-regulatory modules (CRMs) of genes operating downstream of graded Shh signaling and bifunctional Gli proteins in neural patterning. Unexpectedly, we find that Gli activators have a noninstructive role in long-range patterning and cooperate with SoxB1 proteins to facilitate a largely concentration-independent mode of gene activation. Instead, the opposing Gli-repressor gradient is interpreted at transcriptional levels, and, together with CRM-specific repressive input of homeodomain proteins, comprises a repressive network that translates graded Shh signaling into regional gene expression patterns. Moreover, local and long-range interpretation of Shh signaling differs with respect to CRM context sensitivity and Gli-activator dependence, and we propose that these differences provide insight into how morphogen function may have mechanistically evolved from an initially binary inductive event.


Assuntos
Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neurogênese , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Proteína GLI1 em Dedos de Zinco
17.
Genes Dev ; 25(23): 2453-64, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22085726

RESUMO

Pluripotent embryonic stem (ES) cells can generate all cell types, but how cell lineages are initially specified and maintained during development remains largely unknown. Different classes of Sox transcription factors are expressed during neurogenesis and have been assigned important roles from early lineage specification to neuronal differentiation. Here we characterize the genome-wide binding for Sox2, Sox3, and Sox11, which have vital functions in ES cells, neural precursor cells (NPCs), and maturing neurons, respectively. The data demonstrate that Sox factor binding depends on developmental stage-specific constraints and reveal a remarkable sequential binding of Sox proteins to a common set of neural genes. Interestingly, in ES cells, Sox2 preselects for neural lineage-specific genes destined to be bound and activated by Sox3 in NPCs. In NPCs, Sox3 binds genes that are later bound and activated by Sox11 in differentiating neurons. Genes prebound by Sox proteins are associated with a bivalent chromatin signature, which is resolved into a permissive monovalent state upon binding of activating Sox factors. These data indicate that a single key transcription factor family acts sequentially to coordinate neural gene expression from the early lineage specification in pluripotent cells to later stages of neuronal development.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/citologia , Fatores de Transcrição SOX/genética , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Genoma , Camundongos , Neurogênese/genética , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição SOX/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXC/genética , Regulação para Cima
18.
PLoS One ; 6(3): e18454, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21483788

RESUMO

The presence of stem cell characteristics in glioma cells raises the possibility that mechanisms promoting the maintenance and self-renewal of tissue specific stem cells have a similar function in tumor cells. Here we characterized human gliomas of various malignancy grades for the expression of stem cell regulatory proteins. We show that cells in high grade glioma co-express an array of markers defining neural stem cells (NSCs) and that these proteins can fulfill similar functions in tumor cells as in NSCs. However, in contrast to NSCs glioma cells co-express neural proteins together with pluripotent stem cell markers, including the transcription factors Oct4, Sox2, Nanog and Klf4. In line with this finding, in high grade gliomas mesodermal- and endodermal-specific transcription factors were detected together with neural proteins, a combination of lineage markers not normally present in the central nervous system. Persistent presence of pluripotent stem cell traits could only be detected in solid tumors, and observations based on in vitro studies and xenograft transplantations in mice imply that this presence is dependent on the combined activity of intrinsic and extrinsic regulatory cues. Together these results demonstrate a general deregulated expression of neural and pluripotent stem cell traits in malignant human gliomas, and indicate that stem cell regulatory factors may provide significant targets for therapeutic strategies.


Assuntos
Glioma/metabolismo , Glioma/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Células-Tronco Pluripotentes/metabolismo , Animais , Western Blotting , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos SCID , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
19.
Exp Neurol ; 229(1): 65-71, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20974131

RESUMO

Cell therapy aiming at the replacement of degenerated neurons is a very attractive approach. By using an established in vitro organotypic brain stem (BS) slice culture we screen for candidate donor cells, some of them being further functionally assessed in in vivo models of sensorineural hearing loss. Both in vitro and in vivo systems show that implanted cells face challenges of survival, targeted migration, differentiation and functional integration with the host tissue. Low success rates are possibly due to the lack of necessary neurotrophic factors, adhesion molecules and guiding cues. Olfactory ensheathing cells (OECs) have been shown to express a number of neurotrophic factors and to promote axonal growth through cell to cell interactions. In the present study we co-cultured OECs with organotypic BS slice in order to see if OECs can serve as a facilitator when screening candidate donor cells in an organotypic culture setup. Here we show that OECs when co-cultured with the auditory BS slice not only promote neurite outgrowth from the cochlear nucleus (CN) region of the BS slice but also support cells by having BS slice axons growing along their processes. These findings further suggest that OECs may enhance survival and targeted migration of candidate donor cells suitable for cell therapy in vitro and in vivo. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.


Assuntos
Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Neuritos/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Animais , Sobrevivência Celular/fisiologia , Técnicas de Cocultura/métodos , Feminino , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
20.
Development ; 137(3): 437-45, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20081190

RESUMO

Homeodomain (HD) transcription factors and components of the Notch pathway [Delta1 (Dll1), Jagged1 (Jag1) and the Fringe (Fng) proteins] are expressed in distinct progenitor domains along the dorsoventral (DV) axis of the developing spinal cord. However, the internal relationship between these two regulatory pathways has not been established. In this report we show that HD proteins act upstream of Notch signalling. Thus, HD proteins control the spatial distribution of Notch ligands and Fng proteins, whereas perturbation of the Notch pathway does not affect the regional expression of HD proteins. Loss of Dll1 or Jag1 leads to a domain-specific increase of neuronal differentiation but does not affect the establishment of progenitor domain boundaries. Moreover, gain-of-function experiments indicate that the ability of Dll1 and Jag1 to activate Notch is limited to progenitors endogenously expressing the respective ligand. Fng proteins enhance Dll1-activated Notch signalling and block Notch activation mediated by Jag1. This finding, combined with the overlapping expression of Fng with Dll1 but not with Jag1, is likely to explain the domain-specific activity of the Notch ligands. This outcome is opposite to the local regulation of Notch activity in most other systems, including the Drosophila wing, where Fng co-localizes with Jagged/Serrate rather than Dll/Delta, which facilitates Notch signalling at regional boundaries instead of within domains. The regulation of Notch activation in the spinal cord therefore appears to endow specific progenitor populations with a domain-wide autonomy in the control of neurogenesis and prevents any inadequate activation of Notch across progenitor domain boundaries.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurogênese , Receptores Notch/metabolismo , Animais , Padronização Corporal/genética , Embrião de Galinha , Peptídeos e Proteínas de Sinalização Intercelular , Ligantes , Camundongos , Medula Espinal , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA