Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 165(5): 701-721, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36636908

RESUMEN

Neuron-restrictive silencer factor/repressor element 1 (RE1)-silencing transcription factor (NRSF/REST) is a transcriptional repressor of a large cluster of neural genes containing RE1 motifs in their promoter region. NRSF/REST is ubiquitously expressed in non-neuronal cells, including astrocytes, while it is down-regulated during neuronal differentiation. While neuronal NRSF/REST homeostatically regulates intrinsic excitability and synaptic transmission, the role of the high NRSF/REST expression levels in the homeostatic functions of astrocytes is poorly understood. Here, we investigated the functional consequences of NRSF/REST deletion in primary cortical astrocytes derived from NRSF/REST conditional knockout mice (KO). We found that NRSF/REST KO astrocyte displayed a markedly reduced activity of inward rectifying K+ channels subtype 4.1 (Kir4.1) underlying spatial K+ buffering that was associated with a decreased expression and activity of the glutamate transporter-1 (GLT-1) responsible for glutamate uptake by astrocytes. The effects of the impaired astrocyte homeostatic functions on neuronal activity were investigated by co-culturing wild-type hippocampal neurons with NRSF/REST KO astrocytes. Interestingly, neurons experienced increased neuronal excitability at high firing rates associated with decrease after hyperpolarization and increased amplitude of excitatory postsynaptic currents. The data indicate that astrocytic NRSF/REST directly participates in neural circuit homeostasis by regulating intrinsic excitability and excitatory transmission and that dysfunctions of NRSF/REST expression in astrocytes may contribute to the pathogenesis of neurological disorders.


Asunto(s)
Astrocitos , Factores de Transcripción , Ratones , Animales , Factores de Transcripción/genética , Astrocitos/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación de la Expresión Génica
2.
Nucleic Acids Res ; 45(6): 3102-3115, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-27956497

RESUMEN

The dynamic interaction of DNA methylation and transcription factor binding in regulating spatiotemporal gene expression is essential for embryogenesis, but the underlying mechanisms remain understudied. In this study, using mouse models and integration of in vitro and in vivo genetic and epigenetic analyses, we show that the binding of REST (repressor element 1 (RE1) silencing transcription factor; also known as NRSF) to its cognate RE1 sequences is temporally regulated by non-CpG methylation. This process is dependent on DNA methyltransferase 3B (DNMT3B) and leads to suppression of adult cardiac genes in developing hearts. We demonstrate that DNMT3B preferentially mediates non-CpG methylation of REST-targeted genes in the developing heart. Downregulation of DNMT3B results in decreased non-CpG methylation of RE1 sequences, reduced REST occupancy, and consequently release of the transcription suppression during later cardiac development. Together, these findings reveal a critical gene silencing mechanism in developing mammalian hearts that is regulated by the dynamic interaction of DNMT3B-mediated non-CpG methylation and REST binding.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Corazón/embriología , Miocardio/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Ratones , Ratones Endogámicos C57BL , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Unión Proteica , Proteínas Represoras/fisiología , ADN Metiltransferasa 3B
3.
Development ; 142(1): 31-40, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25516968

RESUMEN

The maintenance and control of pluripotency is of great interest in stem cell biology. The dual specificity T-box/basic-helix-loop-helix-zipper transcription factor Mga is expressed in the pluripotent cells of the inner cell mass (ICM) and epiblast of the peri-implantation mouse embryo, but its function has not been investigated previously. Here, we use a loss-of-function allele and RNA knockdown to demonstrate that Mga depletion leads to the death of proliferating pluripotent ICM cells in vivo and in vitro, and the death of embryonic stem cells (ESCs) in vitro. Additionally, quiescent pluripotent cells lacking Mga are lost during embryonic diapause. Expression of Odc1, the rate-limiting enzyme in the conversion of ornithine into putrescine in the synthesis of polyamines, is reduced in Mga mutant cells, and the survival of mutant ICM cells as well as ESCs is rescued in culture by the addition of exogenous putrescine. These results suggest a mechanism whereby Mga influences pluripotent cell survival through regulation of the polyamine pool in pluripotent cells of the embryo, whether they are in a proliferative or quiescent state.


Asunto(s)
Implantación del Embrión , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Alelos , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Masa Celular Interna del Blastocisto/citología , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Cruzamientos Genéticos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Marcación de Gen , Genotipo , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Masculino , Ratones , Mutagénesis/genética , Mutación/genética , Ornitina Descarboxilasa/metabolismo , Células Madre Pluripotentes/metabolismo , Poliaminas/metabolismo , Factores de Transcripción/deficiencia
4.
Hum Mol Genet ; 24(8): 2247-66, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25552653

RESUMEN

Mitochondrial diseases often exhibit tissue-specific pathologies, but this phenomenon is poorly understood. Here we present regulation of mitochondrial translation by the Mitochondrial Translation Optimization Factor 1, MTO1, as a novel player in this scenario. We demonstrate that MTO1 mediates tRNA modification and controls mitochondrial translation rate in a highly tissue-specific manner associated with tissue-specific OXPHOS defects. Activation of mitochondrial proteases, aberrant translation products, as well as defects in OXPHOS complex assembly observed in MTO1 deficient mice further imply that MTO1 impacts translation fidelity. In our mouse model, MTO1-related OXPHOS deficiency can be bypassed by feeding a ketogenic diet. This therapeutic intervention is independent of the MTO1-mediated tRNA modification and involves balancing of mitochondrial and cellular secondary stress responses. Our results thereby establish mammalian MTO1 as a novel factor in the tissue-specific regulation of OXPHOS and fine tuning of mitochondrial translation accuracy.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/metabolismo , Fosforilación Oxidativa , Biosíntesis de Proteínas , ARN de Transferencia/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Dieta Cetogénica , Fibroblastos/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales , Datos de Secuencia Molecular , Especificidad de Órganos , ARN de Transferencia/genética , Proteínas de Unión al ARN , Alineación de Secuencia
5.
Circ Res ; 115(1): 23-31, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24748541

RESUMEN

RATIONALE: 22q11 deletion syndrome arises from recombination between low-copy repeats on chromosome 22. Typical deletions result in hemizygosity for TBX1 associated with congenital cardiovascular disease. Deletions distal to the typically deleted region result in a similar cardiac phenotype but lack in extracardiac features of the syndrome, suggesting that a second haploinsufficient gene maps to this interval. OBJECTIVE: The transcription factor HIC2 is lost in most distal deletions, as well as in a minority of typical deletions. We used mouse models to test the hypothesis that HIC2 hemizygosity causes congenital heart disease. METHODS AND RESULTS: We created a genetrap mouse allele of Hic2. The genetrap reporter was expressed in the heart throughout the key stages of cardiac morphogenesis. Homozygosity for the genetrap allele was embryonic lethal before embryonic day E10.5, whereas the heterozygous condition exhibited a partially penetrant late lethality. One third of heterozygous embryos had a cardiac phenotype. MRI demonstrated a ventricular septal defect with over-riding aorta. Conditional targeting indicated a requirement for Hic2 within the Nkx2.5+ and Mesp1+ cardiovascular progenitor lineages. Microarray analysis revealed increased expression of Bmp10. CONCLUSIONS: Our results demonstrate a novel role for Hic2 in cardiac development. Hic2 is the first gene within the distal 22q11 interval to have a demonstrated haploinsufficient cardiac phenotype in mice. Together our data suggest that HIC2 haploinsufficiency likely contributes to the cardiac defects seen in distal 22q11 deletion syndrome.


Asunto(s)
Síndrome de Deleción 22q11/etiología , Corazón/embriología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Supresoras de Tumor/fisiología , Síndrome de Deleción 22q11/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas Morfogenéticas Óseas/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Cardiopatías Congénitas/etiología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Morfogénesis , Mutagénesis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Proteínas Supresoras de Tumor/genética
6.
J Biol Chem ; 289(15): 10769-10784, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24515116

RESUMEN

The majority of amyotrophic lateral sclerosis (ALS) cases as well as many patients suffering from frontotemporal lobar dementia (FTLD) with ubiquitinated inclusion bodies show TDP-43 pathology, the protein encoded by the TAR DNA-binding protein (Tardbp) gene. We used recombinase-mediated cassette exchange to introduce an ALS patient cDNA into the mouse Tdp-43 locus. Expression levels of human A315T TDP-43 protein were 300% elevated in heterozygotes, whereas the endogenous mouse Tdp-43 was decreased to 20% of wild type levels as a result of disturbed feedback regulation. Heterozygous TDP-43(A315TKi) mutants lost 10% of their body weight and developed insoluble TDP-43 protein starting as early as 3 months after birth, a pathology that was exacerbated with age. We analyzed the splicing patterns of known Tdp-43 target genes as well as genome-wide gene expression levels in different tissues that indicated mitochondrial dysfunction. In heterozygous mutant animals, we observed a relative decrease in expression of Parkin (Park2) and the fatty acid transporter CD36 along with an increase in fatty acids, HDL cholesterol, and glucose in the blood. As seen in transmission electron microscopy, neuronal cells in motor cortices of TDP-43(A315TKi) animals had abnormal neuronal mitochondrial cristae formation. Motor neurons were reduced to 90%, but only slight motoric impairment was detected. The observed phenotype was interpreted as a predisease model, which might be valuable for the identification of further environmental or genetic triggers of neurodegeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Mitocondrias/patología , Alelos , Esclerosis Amiotrófica Lateral/genética , Animales , Conducta Animal , Glucemia/metabolismo , Peso Corporal , Antígenos CD36/metabolismo , HDL-Colesterol/metabolismo , ADN Complementario/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Ácidos Grasos/metabolismo , Femenino , Técnicas de Sustitución del Gen , Genoma , Genotipo , Heterocigoto , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Fenotipo , Ubiquitina-Proteína Ligasas/metabolismo
7.
Nat Med ; 13(3): 324-31, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322895

RESUMEN

In the adult heart, a variety of stresses induce re-expression of a fetal gene program in association with myocyte hypertrophy and heart failure. Here we show that histone deacetylase-2 (Hdac2) regulates expression of many fetal cardiac isoforms. Hdac2 deficiency or chemical histone deacetylase (HDAC) inhibition prevented the re-expression of fetal genes and attenuated cardiac hypertrophy in hearts exposed to hypertrophic stimuli. Resistance to hypertrophy was associated with increased expression of the gene encoding inositol polyphosphate-5-phosphatase f (Inpp5f) resulting in constitutive activation of glycogen synthase kinase 3beta (Gsk3beta) via inactivation of thymoma viral proto-oncogene (Akt) and 3-phosphoinositide-dependent protein kinase-1 (Pdk1). In contrast, Hdac2 transgenic mice had augmented hypertrophy associated with inactivated Gsk3beta. Chemical inhibition of activated Gsk3beta allowed Hdac2-deficient adults to become sensitive to hypertrophic stimulation. These results suggest that Hdac2 is an important molecular target of HDAC inhibitors in the heart and that Hdac2 and Gsk3beta are components of a regulatory pathway providing an attractive therapeutic target for the treatment of cardiac hypertrophy and heart failure.


Asunto(s)
Cardiomegalia/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Histona Desacetilasas/fisiología , Proteínas Represoras/fisiología , Animales , Cardiomegalia/embriología , Cardiomegalia/genética , Activación Enzimática/fisiología , Feto , Glucógeno Sintasa Quinasa 3 beta , Histona Desacetilasa 2 , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transducción de Señal/fisiología
8.
iScience ; 27(4): 109507, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38591011

RESUMEN

Visual cortical plasticity is high during early life, but gradually decreases with development. This is due to the Otx2-driven maturation of intracortical inhibition that parallels the condensation of extracellular matrix components into perineuronal nets mainly around parvalbumin-positive GABAergic neurons. Repressor Element 1 Silencing Transcription (REST) epigenetically controls the expression of a plethora of neuron-specific genes. We demonstrate that the conditional knockout of REST in the primary visual cortex of adult mice induces a shift of ocular dominance after short-term monocular deprivation and promotes the recovery of vision in long-term deprived animals after reverse suture. These phenomena paralleled a reduction of perineuronal net density and increased expression of REST target genes, but not of the homeoprotein Otx2 in the visual cortex contralateral to the deprived eye. This shows that REST regulates adult visual cortical plasticity and is a potential therapeutic target to restore vision in adult amblyopia by enhancing V1 plasticity.

9.
Acta Physiol (Oxf) ; 240(6): e14146, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38606882

RESUMEN

AIM: The Repressor Element-1 Silencing Transcription Factor (REST) is an epigenetic master regulator playing a crucial role in the nervous system. In early developmental stages, REST downregulation promotes neuronal differentiation and the acquisition of the neuronal phenotype. In addition, postnatal fluctuations in REST expression contribute to shaping neuronal networks and maintaining network homeostasis. Here we investigate the role of the early postnatal deletion of neuronal REST in the assembly and strength of excitatory and inhibitory synaptic connections. METHODS: We investigated excitatory and inhibitory synaptic transmission by patch-clamp recordings in acute neocortical slices in a conditional knockout mouse model (RestGTi) in which Rest was deleted by delivering PHP.eB adeno-associated viruses encoding CRE recombinase under the control of the human synapsin I promoter in the lateral ventricles of P0-P1 pups. RESULTS: We show that, under physiological conditions, Rest deletion increased the intrinsic excitability of principal cortical neurons in the primary visual cortex and the density and strength of excitatory synaptic connections impinging on them, without affecting inhibitory transmission. Conversely, in the presence of a pathological excitation/inhibition imbalance induced by pentylenetetrazol, Rest deletion prevented the increase in synaptic excitation and decreased seizure severity. CONCLUSION: The data indicate that REST exerts distinct effects on the excitability of cortical circuits depending on whether it acts under physiological conditions or in the presence of pathologic network hyperexcitability. In the former case, REST preserves a correct excitatory/inhibitory balance in cortical circuits, while in the latter REST loses its homeostatic activity and may become pro-epileptogenic.


Asunto(s)
Corteza Cerebral , Homeostasis , Proteínas Represoras , Animales , Ratones , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Homeostasis/fisiología , Ratones Noqueados , Red Nerviosa/fisiología , Red Nerviosa/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/fisiopatología , Transmisión Sináptica/fisiología
10.
Genome Res ; 20(8): 1154-64, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20548051

RESUMEN

Mutational screens are an effective means used in the functional annotation of a genome. We present a method for a mutational screen of the mouse X chromosome using gene trap technologies. This method has the potential to screen all of the genes on the X chromosome without establishing mutant animals, as all gene-trapped embryonic stem (ES) cell lines are hemizygous null for mutations on the X chromosome. Based on this method, embryonic morphological phenotypes and expression patterns for 58 genes were assessed, approximately 10% of all human and mouse syntenic genes on the X chromosome. Of these, 17 are novel embryonic lethal mutations and nine are mutant mouse models of genes associated with genetic disease in humans, including BCOR and PORCN. The rate of lethal mutations is similar to previous mutagenic screens of the autosomes. Interestingly, some genes associated with X-linked mental retardation (XLMR) in humans show lethal phenotypes in mice, suggesting that null mutations cannot be responsible for all cases of XLMR. The entire data set is available via the publicly accessible website (http://xlinkedgenes.ibme.utoronto.ca/).


Asunto(s)
Análisis Mutacional de ADN/métodos , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Fenotipo , Cromosoma X/genética , Aciltransferasas , Animales , Secuencia de Bases , Cromosomas Humanos X/genética , Genes Letales , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Proteínas de la Membrana/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética
11.
Blood ; 118(3): 554-64, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21613258

RESUMEN

We used a retroviral integration screen to search for novel genes that regulate HSC function. One of the genes that conferred HSC dominance when overexpressed due to an adjacent retroviral insertion was Musashi 2 (Msi2), an RNA-binding protein that can act as a translational inhibitor. A gene-trap mouse model that inactivates the gene shows that Msi2 is more highly expressed in long-term (LT) and short-term (ST) HSCs, as well as in lymphoid myeloid primed progenitors (LMPPs), but much less in intermediate progenitors and mature cells. Mice lacking Msi2 are fully viable for up to a year or more, but exhibit severe defects in primitive precursors, most significantly a reduction in the number of ST-HSCs and LMPPs and a decrease in leukocyte numbers, effects that are exacerbated with age. Cell-cycle and gene-expression analyses suggest that the main hematopoietic defect in Msi2-defective mice is the decreased proliferation capacity of ST-HSCs and LMPPs. In addition, HSCs lacking Msi2 are severely impaired in competitive repopulation experiments, being overgrown by wild-type cells even when mutant cells were provided in excess. Our data indicate that Msi2 maintains the stem cell compartment mainly by regulating the proliferation of primitive progenitors downstream of LT-HSCs.


Asunto(s)
Células Progenitoras Linfoides/fisiología , Células Progenitoras Mieloides/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Adhesión Celular/fisiología , División Celular/fisiología , Movimiento Celular/fisiología , Femenino , Pruebas Genéticas/métodos , Recuento de Leucocitos , Células Progenitoras Linfoides/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Insercional , Células Progenitoras Mieloides/citología , Neomicina , Inhibidores de la Síntesis de la Proteína , Retroviridae/genética , Bazo/patología , Timo/patología , beta-Galactosidasa/genética
12.
Front Cell Neurosci ; 17: 1267609, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38034589

RESUMEN

The repressor element-1 silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is an epigenetic master regulator that plays a crucial role during nervous system development and maturation. REST function was originally described during development, where it determines neuronal phenotype. However, recent studies showed that REST participates in several processes in the adult brain, including neuronal plasticity and epileptogenesis. In this regard, the relationships between REST and epilepsy are still controversial and need further investigation. As forebrain excitatory neurons are the common final pathway of seizure susceptibility, we investigated the role of REST in epilepsy by inducing REST conditional knockout (REST-cKO) specifically in excitatory neurons of the hippocampus. To target the excitatory neuronal population, we cloned the calcium/calmodulin-dependent protein kinase IIα minimal promoter upstream of Cre recombinase. After assessing the specificity of the promoter's expression, the transgenes were packaged in an engineered adeno-associated virus able to cross the blood-brain and blood-cerebrospinal fluid barriers and delivered in the lateral ventricles of 2-month-old RESTflox/flox mice to characterize, after 1 month, the cognitive phenotype and the seizure propensity. We show that REST-cKO mice display lower levels of anxiety in the light-dark test with respect to control mice but have unaltered motor, social, and cognitive profiles. The evaluation of the susceptibility to epileptic seizures showed that REST-cKO mice are more resistant to pentylenetetrazole-induced kindling but not to seizures induced by a single administration of the convulsant and show higher survival rates. Overall, these data suggest that the absence of REST in forebrain excitatory neurons decreases seizure susceptibility, pointing to a pro-epileptogenic role of the transcriptional repressor under conditions of pathological excitation/inhibition imbalance.

13.
Nucleic Acids Res ; 38(9): e106, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20139417

RESUMEN

Recombinase-mediated cassette exchange (RMCE) exploits the possibility to unidirectionally exchange any genetic material flanked by heterotypic recombinase recognition sites (RRS) with target sites in the genome. Due to a limited number of available pre-fabricated target sites, RMCE in mouse embryonic stem (ES) cells has not been tapped to its full potential to date. Here, we introduce a universal system, which allows the targeted insertion of any given transcriptional unit into 85 742 previously annotated retroviral conditional gene trap insertions, representing 7013 independent genes in mouse ES cells, by RMCE. This system can be used to express any given cDNA under the control of endogenous trapped promoters in vivo, as well as for the generation of transposon 'launch pads' for chromosomal region-specific 'Sleeping Beauty' insertional mutagenesis. Moreover, transcription of the gene-of-interest is only activated upon Cre-recombinase activity, a feature that adds conditionality to this expression system, which is demonstrated in vivo. The use of the RMCE system presented in this work requires one single-cloning step followed by one overnight gateway clonase reaction and subsequent cassette exchange in ES cells with efficiencies of 40% in average.


Asunto(s)
ADN Complementario/biosíntesis , Marcación de Gen/métodos , Recombinasas/metabolismo , Animales , Cinamatos/farmacología , ADN Nucleotidiltransferasas/metabolismo , Marcadores Genéticos , Vectores Genéticos , Higromicina B/análogos & derivados , Higromicina B/farmacología , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Regiones Promotoras Genéticas , Recombinación Genética , Transposasas/metabolismo
15.
Aging Cell ; 20(10): e13471, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34520100

RESUMEN

During aging, brain performances decline. Cellular senescence is one of the aging drivers and a key feature of a variety of human age-related disorders. The transcriptional repressor RE1-silencing transcription factor (REST) has been associated with aging and higher risk of neurodegenerative disorders. However, how REST contributes to the senescence program and functional impairment remains largely unknown. Here, we report that REST is essential to prevent the senescence phenotype in primary mouse neurons. REST deficiency causes failure of autophagy and loss of proteostasis, increased oxidative stress, and higher rate of cell death. Re-establishment of autophagy reverses the main hallmarks of senescence. Our data indicate that REST has a protective role in physiological aging by regulating the autophagic flux and the senescence program in neurons, with implications for neurological disorders associated with aging.


Asunto(s)
Autofagia/genética , Senescencia Celular/genética , Neuronas/metabolismo , Proteínas Represoras/deficiencia , Animales , Humanos , Ratones , Estrés Oxidativo
16.
Cell Death Dis ; 12(2): 180, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589593

RESUMEN

Neuroinflammation is associated with synapse dysfunction and cognitive decline in patients and animal models. One candidate for translating the inflammatory stress into structural and functional changes in neural networks is the transcriptional repressor RE1-silencing transcription factor (REST) that regulates the expression of a wide cluster of neuron-specific genes during neurogenesis and in mature neurons. To study the cellular and molecular pathways activated under inflammatory conditions mimicking the experimental autoimmune encephalomyelitis (EAE) environment, we analyzed REST activity in neuroblastoma cells and mouse cortical neurons treated with activated T cell or microglia supernatant and distinct pro-inflammatory cytokines. We found that REST is activated by a variety of neuroinflammatory stimuli in both neuroblastoma cells and primary neurons, indicating that a vast transcriptional change is triggered during neuroinflammation. While a dual activation of REST and its dominant-negative splicing isoform REST4 was observed in N2a neuroblastoma cells, primary neurons responded with a pure full-length REST upregulation in the absence of changes in REST4 expression. In both cases, REST upregulation was associated with activation of Wnt signaling and increased nuclear translocation of ß-catenin, a well-known intracellular transduction pathway in neuroinflammation. Among single cytokines, IL-1ß caused a potent and prompt increase in REST transcription and translation in neurons, which promoted a delayed and strong synaptic downscaling specific for excitatory synapses, with decreased frequency and amplitude of spontaneous synaptic currents, decreased density of excitatory synaptic connections, and decreased frequency of action potential-evoked Ca2+ transients. Most important, the IL-1ß effects on excitatory transmission were strictly REST dependent, as conditional deletion of REST completely occluded the effects of IL-1ß activation on synaptic transmission and network excitability. Our results demonstrate that REST upregulation represents a new pathogenic mechanism for the synaptic dysfunctions observed under neuroinflammatory conditions and identify the REST pathway as therapeutic target for EAE and, potentially, for multiple sclerosis.


Asunto(s)
Corteza Cerebral/metabolismo , Inflamación/metabolismo , Interleucina-1beta/farmacología , Proteínas Represoras/metabolismo , Transmisión Sináptica , Animales , Corteza Cerebral/citología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Proteínas Represoras/biosíntesis , Transmisión Sináptica/efectos de los fármacos , Linfocitos T/metabolismo , Regulación hacia Arriba
18.
Biochim Biophys Acta ; 1787(5): 371-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19111522

RESUMEN

The Tim23 protein is the key component of the mitochondrial import machinery. It locates to the inner mitochondrial membrane and its own import is dependent on the DDP1/TIM13 complex. Mutations in human DDP1 cause the Mohr-Tranebjaerg syndrome (MTS/DFN-1; OMIM #304700), which is one of the two known human diseases of the mitochondrial protein import machinery. We created a Tim23 knockout mouse from a gene trap embryonic stem cell clone. Homozygous Tim23 mice were not viable. Heterozygous F1 mutants showed a 50% reduction of Tim23 protein in Western blot, a neurological phenotype and a markedly reduced life span. Haploinsufficiency of the Tim23 mutation underlines the critical role of the mitochondrial import machinery for maintaining mitochondrial function.


Asunto(s)
Esperanza de Vida , Proteínas de la Membrana/deficiencia , Mitocondrias/genética , Mutación , Animales , Blastocisto/fisiología , Miembro Anterior/fisiología , Genotipo , Fuerza de la Mano , Humanos , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Síndromes Orofaciodigitales/genética , Fenotipo , Transporte de Proteínas/genética , Prueba de Desempeño de Rotación con Aceleración Constante
19.
Biochem J ; 418(1): 61-71, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18973473

RESUMEN

STAF [Sec (selenocysteine) tRNA gene transcription activating factor] is a transcription activating factor for a number of RNA Pol III- and RNA Pol II-dependent genes including the Trsp [Sec tRNA gene], which in turn controls the expression of all selenoproteins. Here, the role of STAF in regulating expression of Sec tRNA and selenoproteins was examined. We generated transgenic mice expressing the Trsp transgene lacking the STAF-binding site and made these mice dependent on the transgene for survival by removing the wild-type Trsp. The level of Sec tRNA was unaffected or slightly elevated in heart and testis, but reduced approximately 60% in liver and kidney, approximately 70% in lung and spleen and approximately 80% in brain and muscle compared with the corresponding organs in control mice. Moreover, the ratio of the two isoforms of Sec tRNA that differ by methylation at position 34 (Um34) was altered significantly, and the Um34-containing form was substantially reduced in all tissues examined. Selenoprotein expression in these animals was most affected in tissues in which the Sec tRNA levels were most severely reduced. Importantly, mice had a neurological phenotype strikingly similar to that of mice in which the selenoprotein P gene had been removed and their life span was substantially reduced. The results indicate that STAF influences selenoprotein expression by enhancing Trsp synthesis in an organ-specific manner and by controlling Sec tRNA modification in each tissue examined.


Asunto(s)
Envejecimiento/fisiología , ARN de Transferencia Aminoácido-Específico/genética , ARN de Transferencia Aminoácido-Específico/metabolismo , Selenoproteínas/metabolismo , Transactivadores/metabolismo , Animales , Encéfalo/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Especificidad de Órganos , Fenotipo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Selenoproteínas/genética , Tasa de Supervivencia , Transactivadores/genética
20.
Nucleic Acids Res ; 36(20): e133, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18812397

RESUMEN

Gene trapping is used to introduce insertional mutations into genes of mouse embryonic stem cells (ESCs). It is performed with gene trap vectors that simultaneously mutate and report the expression of the endogenous gene at the site of insertion and provide a DNA tag for rapid identification of the disrupted gene. Gene traps have been employed worldwide to assemble libraries of mouse ESC lines harboring mutations in single genes, which can be used to make mutant mice. However, most of the employed gene trap vectors require gene expression for reporting a gene trap event and therefore genes that are poorly expressed may be under-represented in the existing libraries. To address this problem, we have developed a novel class of gene trap vectors that can induce gene expression at insertion sites, thereby bypassing the problem of intrinsic poor expression. We show here that the insertion of the osteopontin enhancer into several conventional gene trap vectors significantly increases the gene trapping efficiency in high-throughput screens and facilitates the recovery of poorly expressed genes.


Asunto(s)
Células Madre Embrionarias/metabolismo , Vectores Genéticos , Mutagénesis Insercional/métodos , Animales , Sitios de Unión , Línea Celular , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteopontina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA