Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(2): 400-400.e1, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35063077
2.
Immunity ; 54(2): 225-234.e6, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33476547

RESUMEN

Microglia are activated in many neurological diseases and have been suggested to play an important role in the development of affective disorders including major depression. To investigate how microglial signaling regulates mood, we used bidirectional chemogenetic manipulations of microglial activity in mice. Activation of microglia in the dorsal striatum induced local cytokine expression and a negative affective state characterized by anhedonia and aversion, whereas inactivation of microglia blocked aversion induced by systemic inflammation. Interleukin-6 signaling and cyclooxygenase-1 mediated prostaglandin synthesis in the microglia were critical for the inflammation-induced aversion. Correspondingly, microglial activation led to a prostaglandin-dependent reduction of the excitability of striatal neurons. These findings demonstrate a mechanism by which microglial activation causes negative affect through prostaglandin-dependent modulation of striatal neurons and indicate that interference with this mechanism could milden the depressive symptoms in somatic and psychiatric diseases involving microglial activation.


Asunto(s)
Anhedonia/fisiología , Cuerpo Estriado/inmunología , Depresión/inmunología , Microglía/inmunología , Neuronas/fisiología , Animales , Animales Modificados Genéticamente , Conducta Animal , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inflamación , Interleucina-6/metabolismo , Activación de Macrófagos , Ratones , Inflamación Neurogénica , Prostaglandinas/metabolismo
3.
EMBO J ; 40(9): e106423, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33644903

RESUMEN

Endogenous retroviruses (ERVs) make up a large fraction of mammalian genomes and are thought to contribute to human disease, including brain disorders. In the brain, aberrant activation of ERVs is a potential trigger for an inflammatory response, but mechanistic insight into this phenomenon remains lacking. Using CRISPR/Cas9-based gene disruption of the epigenetic co-repressor protein Trim28, we found a dynamic H3K9me3-dependent regulation of ERVs in proliferating neural progenitor cells (NPCs), but not in adult neurons. In vivo deletion of Trim28 in cortical NPCs during mouse brain development resulted in viable offspring expressing high levels of ERVs in excitatory neurons in the adult brain. Neuronal ERV expression was linked to activated microglia and the presence of ERV-derived proteins in aggregate-like structures. This study demonstrates that brain development is a critical period for the silencing of ERVs and provides causal in vivo evidence demonstrating that transcriptional activation of ERV in neurons results in an inflammatory response.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encefalitis/genética , Retrovirus Endógenos/genética , Eliminación de Gen , Proteína 28 que Contiene Motivos Tripartito/genética , Animales , Encéfalo/inmunología , Encéfalo/virología , Sistemas CRISPR-Cas , Células Cultivadas , Encefalitis/inmunología , Encefalitis/virología , Retrovirus Endógenos/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Activación Transcripcional
4.
Trends Genet ; 36(8): 610-623, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32499105

RESUMEN

The etiology of most neurological disorders is poorly understood and current treatments are largely ineffective. New ideas and concepts are therefore vitally important for future research in this area. This review explores the concept that dysregulation of transposable elements (TEs) contributes to the appearance and pathology of neurodevelopmental and neurodegenerative disorders. Despite TEs making up at least half of the human genome, they are vastly understudied in relation to brain disorders. However, recent advances in sequencing technologies and gene editing approaches are now starting to unravel the pathological role of TEs. Aberrant activation of TEs has been found in many neurological disorders; the resulting pathogenic effects, which include alterations of gene expression, neuroinflammation, and direct neurotoxicity, are starting to be resolved. An increased understanding of the relationship between TEs and pathological processes in the brain improves the potential for novel diagnostics and interventions for brain disorders.


Asunto(s)
Elementos Transponibles de ADN , Evolución Molecular , Genoma Humano , Enfermedades Neurodegenerativas/genética , Trastornos del Neurodesarrollo/genética , Humanos , Enfermedades Neurodegenerativas/patología , Trastornos del Neurodesarrollo/patología
5.
Mol Psychiatry ; 27(8): 3247-3261, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35618883

RESUMEN

Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A2AR), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2AR with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner.


Asunto(s)
Dopamina , Esquizofrenia , Animales , Ratones , Dopamina/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Esquizofrenia/metabolismo , Cuerpo Estriado/metabolismo , Transducción de Señal
6.
Brain ; 145(9): 3035-3057, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34936701

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by CAG expansions in the huntingtin (HTT) gene. Modelling Huntington's disease is challenging, as rodent and cellular models poorly recapitulate the disease as seen in ageing humans. To address this, we generated induced neurons through direct reprogramming of human skin fibroblasts, which retain age-dependent epigenetic characteristics. Huntington's disease induced neurons (HD-iNs) displayed profound deficits in autophagy, characterized by reduced transport of late autophagic structures from the neurites to the soma. These neurite-specific alterations in autophagy resulted in shorter, thinner and fewer neurites specifically in HD-iNs. CRISPRi-mediated silencing of HTT did not rescue this phenotype but rather resulted in additional autophagy alterations in control induced neurons, highlighting the importance of wild-type HTT in normal neuronal autophagy. In summary, our work identifies a distinct subcellular autophagy impairment in adult patient derived Huntington's disease neurons and provides a new rationale for future development of autophagy activation therapies.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Adulto , Autofagia/fisiología , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Neuronas
7.
PLoS Genet ; 15(3): e1008036, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30865625

RESUMEN

Transposable elements (TEs) are dynamically expressed at high levels in multiple human tissues, but the function of TE-derived transcripts remains largely unknown. In this study, we identify numerous TE-derived microRNAs (miRNAs) by conducting Argonaute2 RNA immunoprecipitation followed by small RNA sequencing (AGO2 RIP-seq) on human brain tissue. Many of these miRNAs originated from LINE-2 (L2) elements, which entered the human genome around 100-300 million years ago. L2-miRNAs derived from the 3' end of the L2 consensus sequence and thus shared very similar sequences, indicating that L2-miRNAs could target transcripts with L2s in their 3'UTR. In line with this, many protein-coding genes carried fragments of L2-derived sequences in their 3'UTR: these sequences served as target sites for L2-miRNAs. L2-miRNAs and their targets were generally ubiquitously expressed at low levels in multiple human tissues, suggesting a role for this network in buffering transcriptional levels of housekeeping genes. In addition, we also found evidence that this network is perturbed in glioblastoma. In summary, our findings uncover a TE-based post-transcriptional network that shapes transcriptional regulation in human cells.


Asunto(s)
Elementos Transponibles de ADN , Elementos de Nucleótido Esparcido Largo , MicroARNs/genética , Regiones no Traducidas 3' , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Secuencia de Bases , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , MicroARNs/metabolismo
8.
EMBO J ; 36(10): 1379-1391, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28336683

RESUMEN

During adult neurogenesis, newly formed olfactory bulb (OB) interneurons migrate radially to integrate into specific layers of the OB Despite the importance of this process, the intracellular mechanisms that regulate radial migration remain poorly understood. Here, we find that microRNA (miRNA) let-7 regulates radial migration by modulating autophagy in new-born neurons. Using Argonaute2 immunoprecipitation, we performed global profiling of miRNAs in adult-born OB neurons and identified let-7 as a highly abundant miRNA family. Knockdown of let-7 in migrating neuroblasts prevented radial migration and led to an immature morphology of newly formed interneurons. This phenotype was accompanied by a decrease in autophagic activity. Overexpression of Beclin-1 or TFEB in new-born neurons lacking let-7 resulted in re-activation of autophagy and restored radial migration. Thus, these results reveal a miRNA-dependent link between autophagy and adult neurogenesis with implications for neurodegenerative diseases where these processes are impaired.


Asunto(s)
Autofagia , Movimiento Celular , MicroARNs/metabolismo , Neuronas/fisiología , Animales , Técnicas de Silenciamiento del Gen , Inmunoprecipitación , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética
9.
Mol Cell ; 44(3): 361-72, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22055183

RESUMEN

The maintenance of H3K9 and DNA methylation at imprinting control regions (ICRs) during early embryogenesis is key to the regulation of imprinted genes. Here, we reveal that ZFP57, its cofactor KAP1, and associated effectors bind selectively to the H3K9me3-bearing, DNA-methylated allele of ICRs in ES cells. KAP1 deletion induces a loss of heterochromatin marks at ICRs, whereas deleting ZFP57 or DNMTs leads to ICR DNA demethylation. Accordingly, we find that ZFP57 and KAP1 associated with DNMTs and hemimethylated DNA-binding NP95. Finally, we identify the methylated TGCCGC hexanucleotide as the motif that is recognized by ZFP57 in all ICRs and in several tens of additional loci, several of which are at least ZFP57-dependently methylated in ES cells. These results significantly advance our understanding of imprinting and suggest a general mechanism for the protection of specific loci against the wave of DNA demethylation that affects the mammalian genome during early embryogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Metilación de ADN , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Metilasas de Modificación del ADN/metabolismo , Técnicas de Inactivación de Genes , N-Metiltransferasa de Histona-Lisina , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteína Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito , Ubiquitina-Proteína Ligasas
10.
Development ; 142(18): 3166-77, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26395143

RESUMEN

MicroRNAs (miRNAs) have been implicated in regulating multiple processes during brain development in various species. However, the function of miRNAs in human brain development remains largely unexplored. Here, we provide a comprehensive analysis of miRNA expression of regionalized neural progenitor cells derived from human embryonic stem cells and human foetal brain. We found miR-92b-3p and miR-130b-5p to be specifically associated with neural progenitors and several miRNAs that display both age-specific and region-specific expression patterns. Among these miRNAs, we identified miR-10 to be specifically expressed in the human hindbrain and spinal cord, while being absent from rostral regions. We found that miR-10 regulates a large number of genes enriched for functions including transcription, actin cytoskeleton and ephrin receptor signalling. When overexpressed, miR-10 influences caudalization of human neural progenitor cells. Together, these data confirm a role for miRNAs in establishing different human neural progenitor populations. This dataset also provides a comprehensive resource for future studies investigating the functional role of different miRNAs in human brain development.


Asunto(s)
Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Análisis de Varianza , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Línea Celular , Cromosomas Artificiales Bacterianos , Cartilla de ADN/genética , Citometría de Flujo , Genes Reporteros/genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Lentivirus , MicroARNs/genética , Células-Madre Neurales/fisiología , Reacción en Cadena de la Polimerasa , Reacción en Cadena en Tiempo Real de la Polimerasa , Mapeo Restrictivo , Factores de Transcripción SOXB1/genética
11.
Development ; 141(7): 1580-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24598163

RESUMEN

New neurons, originating from the subventricular zone, are continuously integrating into neuronal circuitry in the olfactory bulb (OB). Using a transgenic sensor mouse, we found that adult-born OB interneurons express microRNA-125 (miR-125), whereas the pre-existing developmentally generated OB interneurons represent a unique population of cells in the adult brain, without miR-125 activity. Stable inhibition of miR-125 in newborn OB neurons resulted in enhanced dendritic morphogenesis, as well as in increased synaptic activation in response to odour sensory stimuli. These data demonstrate that miR-125 controls functional synaptic integration of adult-born OB interneurons. Our results also suggest that absence of an otherwise broadly expressed miRNA is a novel mechanism with which to achieve neuronal subtype specification.


Asunto(s)
Células Madre Adultas/fisiología , Células Madre Embrionarias/fisiología , Interneuronas/fisiología , MicroARNs/fisiología , Bulbo Olfatorio/citología , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Diferenciación Celular/genética , Femenino , Interneuronas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/fisiología , Neurogénesis/genética , Bulbo Olfatorio/embriología , Bulbo Olfatorio/metabolismo , Sinapsis/genética
12.
Genome Res ; 23(3): 452-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23233547

RESUMEN

TRIM28 is critical for the silencing of endogenous retroviruses (ERVs) in embryonic stem (ES) cells. Here, we reveal that an essential impact of this process is the protection of cellular gene expression in early embryos from perturbation by cis-acting activators contained within these retroelements. In TRIM28-depleted ES cells, repressive chromatin marks at ERVs are replaced by histone modifications typical of active enhancers, stimulating transcription of nearby cellular genes, notably those harboring bivalent promoters. Correspondingly, ERV-derived sequences can repress or enhance expression from an adjacent promoter in transgenic embryos depending on their TRIM28 sensitivity in ES cells. TRIM28-mediated control of ERVs is therefore crucial not just to prevent retrotransposition, but more broadly to safeguard the transcriptional dynamics of early embryos.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Retroelementos , Transcripción Genética , Animales , Cromatina/genética , Cromatina/metabolismo , Mapeo Cromosómico , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Sitios Genéticos , Histonas/genética , Histonas/metabolismo , Ratones , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Análisis de Secuencia de ARN , Proteína 28 que Contiene Motivos Tripartito , Regulación hacia Arriba
13.
Nature ; 463(7278): 237-40, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-20075919

RESUMEN

More than forty per cent of the mammalian genome is derived from retroelements, of which about one-quarter are endogenous retroviruses (ERVs). Some are still active, notably in mice the highly polymorphic early transposon (ETn)/MusD and intracisternal A-type particles (IAP). ERVs are transcriptionally silenced during early embryogenesis by histone and DNA methylation (and reviewed in ref. 7), although the initiators of this process, which is essential to protect genome integrity, remain largely unknown. KAP1 (KRAB-associated protein 1, also known as tripartite motif-containing protein 28, TRIM28) represses genes by recruiting the histone methyltransferase SETDB1, heterochromatin protein 1 (HP1) and the NuRD histone deacetylase complex, but few of its physiological targets are known. Two lines of evidence suggest that KAP1-mediated repression could contribute to the control of ERVs: first, KAP1 can trigger permanent gene silencing during early embryogenesis, and second, a KAP1 complex silences the retrovirus murine leukaemia virus in embryonic cells. Consistent with this hypothesis, here we show that KAP1 deletion leads to a marked upregulation of a range of ERVs, in particular IAP elements, in mouse embryonic stem (ES) cells and in early embryos. We further demonstrate that KAP1 acts synergistically with DNA methylation to silence IAP elements, and that it is enriched at the 5' untranslated region (5'UTR) of IAP genomes, where KAP1 deletion leads to the loss of histone 3 lysine 9 trimethylation (H3K9me3), a hallmark of KAP1-mediated repression. Correspondingly, IAP 5'UTR sequences can impose in cis KAP1-dependent repression on a heterologous promoter in ES cells. Our results establish that KAP1 controls endogenous retroelements during early embryonic development.


Asunto(s)
Células Madre Embrionarias/metabolismo , Retrovirus Endógenos/genética , Silenciador del Gen , Genes de Partícula A Intracisternal/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Regiones no Traducidas 5'/genética , Acetilación , Animales , Metilación de ADN , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/virología , Células Madre Embrionarias/virología , Fibroblastos , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/fisiología , Lisina/metabolismo , Metilación , Ratones , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito
14.
Proc Natl Acad Sci U S A ; 110(19): E1817-26, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610405

RESUMEN

The aggregation of α-synuclein plays a major role in Parkinson disease (PD) pathogenesis. Recent evidence suggests that defects in the autophagy-mediated clearance of α-synuclein contribute to the progressive loss of nigral dopamine neurons. Using an in vivo model of α-synuclein toxicity, we show that the PD-like neurodegenerative changes induced by excess cellular levels of α-synuclein in nigral dopamine neurons are closely linked to a progressive decline in markers of lysosome function, accompanied by cytoplasmic retention of transcription factor EB (TFEB), a major transcriptional regulator of the autophagy-lysosome pathway. The changes in lysosomal function, observed in the rat model as well as in human PD midbrain, were reversed by overexpression of TFEB, which afforded robust neuroprotection via the clearance of α-synuclein oligomers, and were aggravated by microRNA-128-mediated repression of TFEB in both A9 and A10 dopamine neurons. Delayed activation of TFEB function through inhibition of mammalian target of rapamycin blocked α-synuclein induced neurodegeneration and further disease progression. The results provide a mechanistic link between α-synuclein toxicity and impaired TFEB function, and highlight TFEB as a key player in the induction of α-synuclein-induced toxicity and PD pathogenesis, thus identifying TFEB as a promising target for therapies aimed at neuroprotection and disease modification in PD.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Mesencéfalo/patología , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Beclina-1 , Encéfalo/metabolismo , Mapeo Encefálico/métodos , Dependovirus , Dopamina , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Lisosomas/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Unión Proteica , Ratas , Ratas Sprague-Dawley
15.
Proc Natl Acad Sci U S A ; 110(17): 7038-43, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23530235

RESUMEN

Cellular reprogramming is a new and rapidly emerging field in which somatic cells can be turned into pluripotent stem cells or other somatic cell types simply by the expression of specific combinations of genes. By viral expression of neural fate determinants, it is possible to directly reprogram mouse and human fibroblasts into functional neurons, also known as induced neurons. The resulting cells are nonproliferating and present an alternative to induced pluripotent stem cells for obtaining patient- and disease-specific neurons to be used for disease modeling and for development of cell therapy. In addition, because the cells do not pass a stem cell intermediate, direct neural conversion has the potential to be performed in vivo. In this study, we show that transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, convert into neurons when reprogramming genes are activated after transplantation. Using a transgenic mouse model to specifically direct expression of reprogramming genes to parenchymal astrocytes residing in the striatum, we also show that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ. Taken together, our data provide proof of principle that direct neural conversion can take place in the adult rodent brain when using transplanted human cells or endogenous mouse cells as a starting cell for neural conversion.


Asunto(s)
Astrocitos/trasplante , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Fibroblastos/trasplante , Neuronas/citología , Animales , Astrocitos/citología , Reprogramación Celular/efectos de los fármacos , Cuerpo Estriado/citología , Doxiciclina/farmacología , Fibroblastos/citología , Citometría de Flujo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Lentivirus , Ratones , Ratones Transgénicos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
16.
Exp Cell Res ; 321(1): 84-9, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24099990

RESUMEN

MicroRNAs (miRNAs) are small, non-coding RNAs that negatively regulate gene expression at the post-transcriptional level. In the brain, a large number of miRNAs are expressed and there is a growing body of evidence demonstrating that miRNAs are essential for brain development and neuronal function. Conditional knockout studies of the core components in the miRNA biogenesis pathway, such as Dicer and DGCR8, have demonstrated a crucial role for miRNAs during the development of the central nervous system. Furthermore, mice deleted for specific miRNAs and miRNA-clusters demonstrate diverse functional roles for different miRNAs during the development of different brain structures. miRNAs have been proposed to regulate cellular functions such as differentiation, proliferation and fate-determination of neural progenitors. In this review we summarise the findings from recent studies that highlight the importance of miRNAs in brain development with a focus on the mouse model. We also discuss the technical limitations of current miRNA studies that still limit our understanding of this family of non-coding RNAs and propose the use of novel and refined technologies that are needed in order to fully determine the impact of specific miRNAs in brain development.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Animales , Ratones
17.
Neuroscientist ; 20(3): 235-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23877999

RESUMEN

Since the discovery of short, regulatory microRNAs (miRNA) 20 years ago, the understanding of their impact on gene regulation has dramatically increased. Differentiation of cells requires comprehensive changes in regulatory networks at all levels of gene expression. Posttranscriptional regulation by miRNA leads to rapid modifications in the protein level of large gene networks, and it is therefore not surprising that miRNAs have been found to influence the fate of differentiating cells. Several recent studies have shown that overexpression of a single miRNA in different cellular contexts results in forced differentiation of nerve cells. Loss of this miRNA constrains neurogenesis and promotes gliogenesis. This miRNA, miR-124, is probably the most well-documented example of a miRNA that controls nerve cell fate determination. In this review we summarize the recent findings on miR-124, potential molecular mechanisms used by miR-124 to drive neuronal differentiation, and outline future directions.


Asunto(s)
Regulación de la Expresión Génica/fisiología , MicroARNs/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Humanos
18.
Proc Natl Acad Sci U S A ; 108(25): 10343-8, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21646515

RESUMEN

Recent reports demonstrate that somatic mouse cells can be directly converted to other mature cell types by using combined expression of defined factors. Here we show that the same strategy can be applied to human embryonic and postnatal fibroblasts. By overexpression of the transcription factors Ascl1, Brn2, and Myt1l, human fibroblasts were efficiently converted to functional neurons. We also demonstrate that the converted neurons can be directed toward distinct functional neurotransmitter phenotypes when the appropriate transcriptional cues are provided together with the three conversion factors. By combining expression of the three conversion factors with expression of two genes involved in dopamine neuron generation, Lmx1a and FoxA2, we could direct the phenotype of the converted cells toward dopaminergic neurons. Such subtype-specific induced neurons derived from human somatic cells could be valuable for disease modeling and cell replacement therapy.


Asunto(s)
Transdiferenciación Celular/fisiología , Dopamina/metabolismo , Fibroblastos/fisiología , Neuronas/fisiología , Potenciales de Acción/fisiología , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Neuronas/citología , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
BMJ Open ; 14(5): e084296, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38803267

RESUMEN

INTRODUCTION: Chronic obstructive pulmonary disease (COPD) is a major global health concern, characterised by ventilatory constraints, decreased cardiovascular fitness and reduced limb muscle function, profoundly affecting patients' quality of life. Aerobic training plays a crucial role in the treatment of COPD, but the variability in methodologies and incomplete reporting of key components in aerobic training trials limits the assessment of their effectiveness. This systematic review aims to critically evaluate the application of training principles and reporting of key components in aerobic training trials in randomised controlled trials (RCTs) in the COPD literature. METHODS AND ANALYSIS: The protocol adheres to the Preferred Reporting Items for Systematic reviews and Meta-Analyses Protocol guidelines. The review will include RCTs utilising aerobic training in individuals with COPD. A comprehensive search, following a predefined search strategy will identify studies published from 2007 to 2024 in English from MEDLINE, Embase, CINAHL, CENTRAL and PEDro. Studies including people with COPD and any aerobic training intervention will be included. Two reviewers will independently screen abstracts and titles for inclusion. Two reviewers will independently conduct the screening of full-text documents and data extraction. Study quality will be assessed using the Tool for the assESsment of sTudy quality and bias in Exercise, specifically developed for exercise training studies. The certainty of the evidence will be evaluated using the Grading of Recommendations Assessment, Development and Evaluation approach. A systematic synthesis will be provided, with meta-analyses and meta-regression when appropriate. ETHICS AND DISSEMINATION: As this review will involve the analysis of published data, ethical approval is not required. The findings of this review will be disseminated through peer-reviewed publications and conference presentations. PROSPERO REGISTRATION NUMBER: CRD42021247343.


Asunto(s)
Metaanálisis como Asunto , Enfermedad Pulmonar Obstructiva Crónica , Revisiones Sistemáticas como Asunto , Humanos , Enfermedad Pulmonar Obstructiva Crónica/terapia , Enfermedad Pulmonar Obstructiva Crónica/rehabilitación , Proyectos de Investigación , Terapia por Ejercicio/métodos , Calidad de Vida , Ejercicio Físico , Ensayos Clínicos Controlados Aleatorios como Asunto
20.
Nat Struct Mol Biol ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834915

RESUMEN

SVA (SINE (short interspersed nuclear element)-VNTR (variable number of tandem repeats)-Alu) retrotransposons remain active in humans and contribute to individual genetic variation. Polymorphic SVA alleles harbor gene regulatory potential and can cause genetic disease. However, how SVA insertions are controlled and functionally impact human disease is unknown. Here we dissect the epigenetic regulation and influence of SVAs in cellular models of X-linked dystonia parkinsonism (XDP), a neurodegenerative disorder caused by an SVA insertion at the TAF1 locus. We demonstrate that the KRAB zinc finger protein ZNF91 establishes H3K9me3 and DNA methylation over SVAs, including polymorphic alleles, in human neural progenitor cells. The resulting mini-heterochromatin domains attenuate the cis-regulatory impact of SVAs. This is critical for XDP pathology; removal of local heterochromatin severely aggravates the XDP molecular phenotype, resulting in increased TAF1 intron retention and reduced expression. Our results provide unique mechanistic insights into how human polymorphic transposon insertions are recognized and how their regulatory impact is constrained by an innate epigenetic defense system.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA