Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 84(4): 611-613, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38364777

RESUMEN

We spoke with co-corresponding authors Nico Wahl, Georg Dechant, and Galina Apostolova about their paper "SATB2 organizes the 3D genome architecture of cognition in cortical neurons" (this issue of Molecular Cell), their paths to a career in science, and the importance of perseverance.


Asunto(s)
Neuronas , Factores de Transcripción , Factores de Transcripción/genética
2.
Mol Cell ; 84(4): 621-639.e9, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38244545

RESUMEN

The DNA-binding protein SATB2 is genetically linked to human intelligence. We studied its influence on the three-dimensional (3D) epigenome by mapping chromatin interactions and accessibility in control versus SATB2-deficient cortical neurons. We find that SATB2 affects the chromatin looping between enhancers and promoters of neuronal-activity-regulated genes, thus influencing their expression. It also alters A/B compartments, topologically associating domains, and frequently interacting regions. Genes linked to SATB2-dependent 3D genome changes are implicated in highly specialized neuronal functions and contribute to cognitive ability and risk for neuropsychiatric and neurodevelopmental disorders. Non-coding DNA regions with a SATB2-dependent structure are enriched for common variants associated with educational attainment, intelligence, and schizophrenia. Our data establish SATB2 as a cell-type-specific 3D genome modulator, which operates both independently and in cooperation with CCCTC-binding factor (CTCF) to set up the chromatin landscape of pyramidal neurons for cognitive processes.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neuronas/metabolismo , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Cromatina/metabolismo , Genoma , Cognición , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo
3.
Hum Mol Genet ; 32(13): 2241-2250, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37027192

RESUMEN

OBJECTIVE: In Friedreich's ataxia (FRDA), the most affected tissues are not accessible to sampling and available transcriptomic findings originate from blood-derived cells and animal models. Herein, we aimed at dissecting for the first time the pathophysiology of FRDA by means of RNA-sequencing in an affected tissue sampled in vivo. METHODS: Skeletal muscle biopsies were collected from seven FRDA patients before and after treatment with recombinant human Erythropoietin (rhuEPO) within a clinical trial. Total RNA extraction, 3'-mRNA library preparation and sequencing were performed according to standard procedures. We tested for differential gene expression with DESeq2 and performed gene set enrichment analysis with respect to control subjects. RESULTS: FRDA transcriptomes showed 1873 genes differentially expressed from controls. Two main signatures emerged: (1) a global downregulation of the mitochondrial transcriptome as well as of ribosome/translational machinery and (2) an upregulation of genes related to transcription and chromatin regulation, especially of repressor terms. Downregulation of the mitochondrial transcriptome was more profound than previously shown in other cellular systems. Furthermore, we observed in FRDA patients a marked upregulation of leptin, the master regulator of energy homeostasis. RhuEPO treatment further enhanced leptin expression. INTERPRETATION: Our findings reflect a double hit in the pathophysiology of FRDA: a transcriptional/translational issue and a profound mitochondrial failure downstream. Leptin upregulation in the skeletal muscle in FRDA may represent a compensatory mechanism of mitochondrial dysfunction, which is amenable to pharmacological boosting. Skeletal muscle transcriptomics is a valuable biomarker to monitor therapeutic interventions in FRDA.


Asunto(s)
Eritropoyetina , Ataxia de Friedreich , Animales , Humanos , Transcriptoma/genética , Leptina/genética , Ataxia de Friedreich/patología , Eritropoyetina/genética , ARN , Músculo Esquelético/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo
4.
Cells ; 11(6)2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35326417

RESUMEN

The induction of lineage-specific gene programs are strongly influenced by alterations in local chromatin architecture. However, key players that impact this genome reorganization remain largely unknown. Here, we report that the removal of the special AT-rich binding protein 2 (SATB2), a nuclear protein known to bind matrix attachment regions, is a key event in initiating myogenic differentiation. The deletion of myoblast SATB2 in vitro initiates chromatin remodeling and accelerates differentiation, which is dependent on the caspase 7-mediated cleavage of SATB2. A genome-wide analysis indicates that SATB2 binding within chromatin loops and near anchor points influences both loop and sub-TAD domain formation. Consequently, the chromatin changes that occur with the removal of SATB2 lead to the derepression of differentiation-inducing factors while also limiting the expression of genes that inhibit this cell fate change. Taken together, this study demonstrates that the temporal control of the SATB2 protein is critical in shaping the chromatin environment and coordinating the myogenic differentiation program.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Caspasas , Cromatina , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Mioblastos/metabolismo , Factores de Transcripción/metabolismo
5.
Nature ; 599(7886): 684-691, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34789882

RESUMEN

The three-dimensional (3D) structure of chromatin is intrinsically associated with gene regulation and cell function1-3. Methods based on chromatin conformation capture have mapped chromatin structures in neuronal systems such as in vitro differentiated neurons, neurons isolated through fluorescence-activated cell sorting from cortical tissues pooled from different animals and from dissociated whole hippocampi4-6. However, changes in chromatin organization captured by imaging, such as the relocation of Bdnf away from the nuclear periphery after activation7, are invisible with such approaches8. Here we developed immunoGAM, an extension of genome architecture mapping (GAM)2,9, to map 3D chromatin topology genome-wide in specific brain cell types, without tissue disruption, from single animals. GAM is a ligation-free technology that maps genome topology by sequencing the DNA content from thin (about 220 nm) nuclear cryosections. Chromatin interactions are identified from the increased probability of co-segregation of contacting loci across a collection of nuclear slices. ImmunoGAM expands the scope of GAM to enable the selection of specific cell types using low cell numbers (approximately 1,000 cells) within a complex tissue and avoids tissue dissociation2,10. We report cell-type specialized 3D chromatin structures at multiple genomic scales that relate to patterns of gene expression. We discover extensive 'melting' of long genes when they are highly expressed and/or have high chromatin accessibility. The contacts most specific of neuron subtypes contain genes associated with specialized processes, such as addiction and synaptic plasticity, which harbour putative binding sites for neuronal transcription factors within accessible chromatin regions. Moreover, sensory receptor genes are preferentially found in heterochromatic compartments in brain cells, which establish strong contacts across tens of megabases. Our results demonstrate that highly specific chromatin conformations in brain cells are tightly related to gene regulation mechanisms and specialized functions.


Asunto(s)
Encéfalo/citología , Células/clasificación , Ensamble y Desensamble de Cromatina , Cromatina/química , Cromatina/genética , Genes , Conformación Molecular , Animales , Sitios de Unión , Células/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Familia de Multigenes/genética , Neuronas/clasificación , Neuronas/metabolismo , Desnaturalización de Ácido Nucleico , Factores de Transcripción/metabolismo
6.
EMBO J ; 40(3): e103701, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33319920

RESUMEN

SATB2 is a schizophrenia risk gene and is genetically associated with human intelligence. How it affects cognition at molecular level is currently unknown. Here, we show that interactions between SATB2, a chromosomal scaffolding protein, and the inner nuclear membrane protein LEMD2 orchestrate the response of pyramidal neurons to neuronal activation. Exposure to novel environment in vivo causes changes in nuclear shape of CA1 hippocampal neurons via a SATB2-dependent mechanism. The activity-driven plasticity of the nuclear envelope requires not only SATB2, but also its protein interactor LEMD2 and the ESCRT-III/VPS4 membrane-remodeling complex. Furthermore, LEMD2 depletion in cortical neurons, similar to SATB2 ablation, affects neuronal activity-dependent regulation of multiple rapid and delayed primary response genes. In human genetic data, LEMD2-regulated genes are enriched for de novo mutations reported in intellectual disability and schizophrenia and are, like SATB2-regulated genes, enriched for common variants associated with schizophrenia and cognitive function. Hence, interactions between SATB2 and the inner nuclear membrane protein LEMD2 influence gene expression programs in pyramidal neurons that are linked to cognitive ability and psychiatric disorder etiology.


Asunto(s)
Redes Reguladoras de Genes , Hipocampo/citología , Discapacidad Intelectual/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Esquizofrenia/genética , Factores de Transcripción/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Núcleo Celular/metabolismo , Plasticidad de la Célula , Células Cultivadas , Cognición , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células HeLa , Hipocampo/metabolismo , Humanos , Discapacidad Intelectual/metabolismo , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/química , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Neuronas/citología , Neuronas/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Esquizofrenia/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo
7.
PLoS Genet ; 15(2): e1007890, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30726206

RESUMEN

During CNS development, the nuclear protein SATB2 is expressed in superficial cortical layers and determines projection neuron identity. In the adult CNS, SATB2 is expressed in pyramidal neurons of all cortical layers and is a regulator of synaptic plasticity and long-term memory. Common variation in SATB2 locus confers risk of schizophrenia, whereas rare, de novo structural and single nucleotide variants cause severe intellectual disability and absent or limited speech. To characterize differences in SATB2 molecular function in developing vs adult neocortex, we isolated SATB2 protein interactomes at the two ontogenetic stages and identified multiple novel SATB2 interactors. SATB2 interactomes are highly enriched for proteins that stabilize de novo chromatin loops. The comparison between the neonatal and adult SATB2 protein complexes indicates a developmental shift in SATB2 molecular function, from transcriptional repression towards organization of chromosomal superstructure. Accordingly, gene sets regulated by SATB2 in the neocortex of neonatal and adult mice show limited overlap. Genes encoding SATB2 protein interactors were grouped for gene set analysis of human GWAS data. Common variants associated with human cognitive ability are enriched within the genes encoding adult but not neonatal SATB2 interactors. Our data support a shift in the function of SATB2 in cortex over lifetime and indicate that regulation of spatial chromatin architecture by the SATB2 interactome contributes to cognitive function in the general population.


Asunto(s)
Cognición/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Neocórtex/fisiología , Factores de Transcripción/genética , Adulto , Animales , Humanos , Memoria a Largo Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Polimorfismo de Nucleótido Simple/genética , Transcripción Genética/genética
8.
Front Behav Neurosci ; 12: 224, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356831

RESUMEN

SATB2 is a DNA binding protein that specifically binds the nuclear matrix attachment region and functions as a regulator of the transcription of large chromatin domains. Unlike its well addressed role during brain development, the role of SATB2 in adult brain is under-investigated. It has been shown that deletion of SATB2 from the forebrain of adult mice significantly impaired long-term memory for contextual fear and object recognition memory. The aim of the present study was to investigate the effects of appetitive stimuli such as cocaine and social interaction (SI) on SATB2 expression in the adult rat brain. For that, we performed conditioned place preference (CPP) to cocaine (15 mg/kg) and to SI, then assessed SATB2 expression in the brain 1 h (24 h after the last conditioning) and 24 h (48 h after the last conditioning) after the CPP test. We found that SATB2 expression in the paraventricular thalamus of rats was increased 1 h after the cocaine CPP test. This increase was selective for the cocaine-paired environment since the SI-paired environment did not increase SATB2 expression in the paraventricular thalamus. Also, the cocaine paired environment-induced increase of SATB2 levels in the paraventricular thalamus was due to cocaine conditioning as the unpaired cocaine group did not show an increase of SATB2 in the paraventricular thalamus. These results suggest that SATB2 in the paraventricular thalamus appears to be involved in the association between cocaine effects and environmental context. Further studies are needed to address the functional role of SATB2 in cocaine conditioning.

9.
PLoS Genet ; 14(7): e1007515, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30040823

RESUMEN

SATB2 is associated with schizophrenia and is an important transcription factor regulating neocortical organization and circuitry. Rare mutations in SATB2 cause a syndrome that includes developmental delay, and mouse studies identify an important role for SATB2 in learning and memory. Interacting partners BCL11B and GATAD2A are also schizophrenia risk genes indicating that other genes interacting with or are regulated by SATB2 are making a contribution to schizophrenia and cognition. We used data from Satb2 mouse models to generate three gene-sets that contain genes either functionally related to SATB2 or targeted by SATB2 at different stages of development. Each was tested for enrichment using the largest available genome-wide association studies (GWAS) datasets for schizophrenia and educational attainment (EA) and enrichment analysis was also performed for schizophrenia and other neurodevelopmental disorders using data from rare variant sequencing studies. These SATB2 gene-sets were enriched for genes containing common variants associated with schizophrenia and EA, and were enriched for genes containing rare variants reported in studies of schizophrenia, autism and intellectual disability. In the developing cortex, genes targeted by SATB2 based on ChIP-seq data, and functionally affected when SATB2 is not expressed based on differential expression analysis using RNA-seq data, show strong enrichment for genes associated with EA. For genes expressed in the hippocampus or at the synapse, those targeted by SATB2 are more strongly enriched for genes associated EA than gene-sets not targeted by SATB2. This study demonstrates that single gene findings from GWAS can provide important insights to pathobiological processes. In this case we find evidence that genes influenced by SATB2 and involved in synaptic transmission, axon guidance and formation of the corpus callosum are contributing to schizophrenia and cognition.


Asunto(s)
Cognición , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Trastornos del Neurodesarrollo/genética , Esquizofrenia/genética , Factores de Transcripción/metabolismo , Éxito Académico , Animales , Orientación del Axón/genética , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/metabolismo , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Genómica/métodos , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Mutación , Trastornos del Neurodesarrollo/patología , Esquizofrenia/patología , Transmisión Sináptica/genética , Factores de Transcripción/genética
10.
Elife ; 52016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27897969

RESUMEN

SATB2 is a risk locus for schizophrenia and encodes a DNA-binding protein that regulates higher-order chromatin configuration. In the adult brain Satb2 is almost exclusively expressed in pyramidal neurons of two brain regions important for memory formation, the cerebral cortex and the CA1-hippocampal field. Here we show that Satb2 is required for key hippocampal functions since deletion of Satb2 from the adult mouse forebrain prevents the stabilization of synaptic long-term potentiation and markedly impairs long-term fear and object discrimination memory. At the molecular level, we find that synaptic activity and BDNF up-regulate Satb2, which itself binds to the promoters of coding and non-coding genes. Satb2 controls the hippocampal levels of a large cohort of miRNAs, many of which are implicated in synaptic plasticity and memory formation. Together, our findings demonstrate that Satb2 is critically involved in long-term plasticity processes in the adult forebrain that underlie the consolidation and stabilization of context-linked memory.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Memoria a Largo Plazo , MicroARNs/biosíntesis , Factores de Transcripción/metabolismo , Animales , Técnicas de Inactivación de Genes , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
11.
Stem Cells ; 33(2): 574-88, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25331182

RESUMEN

Neural crest-derived stem cells (NCSCs) from the embryonic peripheral nervous system (PNS) can be reprogrammed in neurosphere (NS) culture to rNCSCs that produce central nervous system (CNS) progeny, including myelinating oligodendrocytes. Using global gene expression analysis we now demonstrate that rNCSCs completely lose their previous PNS characteristics and acquire the identity of neural stem cells derived from embryonic spinal cord. Reprogramming proceeds rapidly and results in a homogenous population of Olig2-, Sox3-, and Lex-positive CNS stem cells. Low-level expression of pluripotency inducing genes Oct4, Nanog, and Klf4 argues against a transient pluripotent state during reprogramming. The acquisition of CNS properties is prevented in the presence of BMP4 (BMP NCSCs) as shown by marker gene expression and the potential to produce PNS neurons and glia. In addition, genes characteristic for mesenchymal and perivascular progenitors are expressed, which suggests that BMP NCSCs are directed toward a pericyte progenitor/mesenchymal stem cell (MSC) fate. Adult NCSCs from mouse palate, an easily accessible source of adult NCSCs, display strikingly similar properties. They do not generate cells with CNS characteristics but lose the neural crest markers Sox10 and p75 and produce MSC-like cells. These findings show that embryonic NCSCs acquire a full CNS identity in NS culture. In contrast, MSC-like cells are generated from BMP NCSCs and pNCSCs, which reveals that postmigratory NCSCs are a source for MSC-like cells up to the adult stage.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Embrión de Mamíferos/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/metabolismo , Médula Espinal/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Factor 4 Similar a Kruppel , Ratones , Cresta Neural/citología , Cresta Neural/embriología , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Médula Espinal/citología , Médula Espinal/embriología
12.
Nucleic Acids Res ; 40(13): 6001-15, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492625

RESUMEN

Protein-coding genes, guiding differentiation of ES cells into neural cells, have extensively been studied in the past. However, for the class of ncRNAs only the involvement of some specific microRNAs (miRNAs) has been described. Thus, to characterize the entire small non-coding RNA (ncRNA) transcriptome, involved in the differentiation of mouse ES cells into neural cells, we have generated three specialized ribonucleo-protein particle (RNP)-derived cDNA libraries, i.e. from pluripotent ES cells, neural progenitors and differentiated neural cells, respectively. By high-throughput sequencing and transcriptional profiling we identified several novel miRNAs to be involved in ES cell differentiation, as well as seven small nucleolar RNAs. In addition, expression of 7SL, 7SK and vault-2 RNAs was significantly up-regulated during ES cell differentiation. About half of ncRNA sequences from the three cDNA libraries mapped to intergenic or intragenic regions, designated as interRNAs and intraRNAs, respectively. Thereby, novel ncRNA candidates exhibited a predominant size of 18-30 nt, thus resembling miRNA species, but, with few exceptions, lacking canonical miRNA features. Additionally, these novel intraRNAs and interRNAs were not only found to be differentially expressed in stem-cell derivatives, but also in primary cultures of hippocampal neurons and astrocytes, strengthening their potential function in neural ES cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Células-Madre Neurales/metabolismo , ARN no Traducido/metabolismo , Animales , Astrocitos/metabolismo , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Biblioteca de Genes , Hipocampo/citología , Hipocampo/metabolismo , Ratones , MicroARNs/metabolismo , Células-Madre Neurales/citología , Neuronas/metabolismo , ARN no Traducido/química , Ribonucleoproteínas/metabolismo
13.
J Neurosci ; 31(34): 12059-67, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21865449

RESUMEN

Although the p38 mitogen-activated protein kinases are active in many neuronal populations in the peripheral and central nervous systems, little is known about the physiological functions of p38 in postmitotic neurons. We report that p38 activity determines in vitro and in vivo the switch from noradrenergic to cholinergic neurotransmission that occurs in sympathetic neurons on exposure to the neuropoietic cytokines CNTF and LIF. This transdifferentiation serves as a model for the plastic mechanisms that enable mature neurons to change some of their central functions without passing through the cell cycle. We demonstrate that in postmitotic neurons, p38 and STAT pathways are concurrently activated by neuropoietic cytokine treatment for at least 12 h overlapping with changes in neurotransmitter marker gene expression. Inhibition of p38 blocks the upregulation of the nuclear matrix protein Satb2 and of cholinergic markers by CNTF without affecting STAT3 phosphorylation. Conversely, overexpression of p38α or ß in the absence of cytokines stimulates cholinergic marker expression. The neurotransmitter switch in vitro is impaired in neurons isolated from p38ß(-/-) mice. Consistent with these in vitro results, a substantial loss of cells expressing cholinergic properties is observed in vivo in the stellate ganglion of mature mice deficient in the p38ß isoform.


Asunto(s)
Acetilcolina/fisiología , Transdiferenciación Celular/genética , Neuronas Colinérgicas/enzimología , Proteína Quinasa 11 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Ganglio Estrellado/enzimología , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Neuronas Colinérgicas/citología , Neuronas Colinérgicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 11 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Neurotransmisores/genética , Neurotransmisores/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ganglio Estrellado/citología , Ganglio Estrellado/crecimiento & desarrollo
14.
J Neurosci ; 30(48): 16356-64, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123581

RESUMEN

Sympathetic neurons can switch their neurotransmitter phenotype from noradrenergic to cholinergic on exposure to neuropoietic cytokines in vitro and in vivo. Here, we provide evidence that this transspecification is regulated by the chromatin architecture protein Satb2. Treatment with the neuropoietic cytokines ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor rapidly and strongly increases Satb2 transcript and protein levels in cultures of rat superior cervical ganglia neurons. Knockdown of endogenous Satb2 by short interfering RNA prevents the upregulation of choline acetyltransferase (Chat) and vesicular acetylcholine transporter (Vacht) by CNTF as well as the loss of norepinephrine transporter (Net). Conversely, overexpression of Satb2 in the noradrenergic sympathetic phenotype results in a marked increase of Chat and Vacht expression and reduced Net mRNA levels in the absence of neuropoietic cytokines. Chromatin immunoprecipitation analysis in primary sympathetic neurons reveals that Satb2 binds to matrix attachment regions (MARs) within the Chat locus. In vivo, in the rat stellate ganglion, Satb2 is expressed exclusively in sudomotor cholinergic neurons innervating the sweat glands and only after establishment of contact between neurons and target. These findings demonstrate a function of the MAR-binding protein Satb2 in growth factor-dependent neurotransmitter plasticity in postmitotic neurons.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Neurotransmisores/fisiología , Ganglio Cervical Superior/fisiología , Factores de Transcripción/fisiología , Fibras Adrenérgicas/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Técnicas de Silenciamiento del Gen , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Mitosis/genética , Mitosis/fisiología , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Neurotransmisores/genética , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/fisiología , Ratas , Factores de Transcripción/genética
15.
Auton Neurosci ; 151(1): 30-8, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19734109

RESUMEN

This review summarizes the current understanding of neurotransmitter phenotype specification of postganglionic sympathetic neurons, focusing, in particular, on the cellular processes of induction versus trans-differentiation. The emerging evidence is discussed that the noradrenergic and cholinergic neurotransmitter phenotypes are co-induced during early development and that the mature phenotypes develop by positive and negative selection of cellular properties in initially bimodal neurons, depending on extracellular signals during migration and after target contact.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Neurotransmisores/biosíntesis , Sistema Nervioso Simpático/citología , Fibras Adrenérgicas/metabolismo , Animales , Transdiferenciación Celular , Embrión de Pollo , Fibras Colinérgicas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Modelos Neurológicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Cresta Neural/citología , Cresta Neural/metabolismo , Neurogénesis , Neuronas/citología , Neurotransmisores/genética , Fenotipo , Proteínas Proto-Oncogénicas c-ret/deficiencia , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/fisiología , Transducción de Señal , Sistema Nervioso Simpático/embriología , Factores de Transcripción/fisiología , Transcripción Genética
16.
Mol Cell Neurosci ; 35(3): 397-408, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17513123

RESUMEN

During late developmental phases individual sympathetic neurons undergo a switch from noradrenergic to cholinergic neurotransmission. This phenomenon of plasticity depends on target-derived signals in vivo and is triggered by neurotrophic factors in neuronal cultures. To analyze genome-wide expression differences between the two transmitter phenotypes we employed DNA microarrays. RNA expression profiles were obtained from chick paravertebral sympathetic ganglia, treated with neurotrophin 3, glial cell line-derived neurotrophic factor or ciliary neurotrophic factor, all of which stimulate cholinergic differentiation. Results were compared with the effect of nerve growth factor, which functions as a pro-noradrenergic stimulus. The gene set common to all three comparisons defined the noradrenergic and cholinergic synexpression groups. Several functional categories, such as signal transduction, G-protein-coupled signaling, cation transport, neurogenesis and synaptic transmission, were enriched in these groups. Experiments based on the prediction that some of the identified genes play a role in the neurotransmitter switch identified bone morphogenetic protein signaling as an inhibitor of cholinergic differentiation.


Asunto(s)
Ganglios Simpáticos/citología , Ganglios Simpáticos/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/fisiología , Neurotransmisores/metabolismo , Fenotipo , Animales , Embrión de Pollo , Perfilación de la Expresión Génica/métodos , Hibridación in Situ/métodos , Neurotransmisores/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Técnicas de Cultivo de Órganos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
17.
Am J Physiol Endocrinol Metab ; 288(5): E957-64, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15613680

RESUMEN

Dehydroepiandrosterone (DHEA) exerts beneficial effects on blood glucose levels and insulin sensitivity in obese rodents and humans, resembling the effects of peroxisome proliferator-activated receptor-gamma (PPARgamma) ligands and opposing those of glucocorticoids; however, the underlying mechanisms remain unclear. Glucocorticoids are reactivated locally by 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which is currently considered as a promising target for the treatment of obesity and diabetes. Using differentiated 3T3-L1 adipocytes, we show that DHEA causes downregulation of 11beta-HSD1 and dose-dependent reduction of its oxoreductase activity. The effects of DHEA were comparable with those of the PPARgamma agonist rosiglitazone but not additive. Furthermore, DHEA reduced the expression of hexose-6-phosphate dehydrogenase, which stimulates the oxoreductase activity of 11beta-HSD1. These findings were confirmed in white adipose tissue and in liver from DHEA-treated C57BL/6J mice. Analysis of the transcription factors involved in the DHEA-dependent regulation of 11beta-HSD1 expression revealed a switch in CCAAT/enhancer-binding protein (C/EBP) expression. C/EBPalpha, a potent activator of 11beta-HSD1 gene transcription, was downregulated in 3T3-L1 adipocytes and in liver and adipose tissue of DHEA-treated mice, whereas C/EBPbeta and C/EBPdelta, attenuating the effect of C/EBPalpha, were unchanged or elevated. Our results further suggest a protective effect of DHEA on adipose tissue by upregulating PPARalpha and downregulating leptin, thereby contributing to the reduced expression of 11beta-HSD1. In summary, we provide evidence that some of the anti-diabetic effects of DHEA may be caused through inhibition of the local amplification of glucocorticoids by 11beta-HSD1 in adipose tissue.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Deshidroepiandrosterona/administración & dosificación , Glucocorticoides/metabolismo , Hígado/metabolismo , Células 3T3-L1 , Animales , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Rosiglitazona , Tiazolidinedionas/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA