Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 143(14): 1399-1413, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38194688

RESUMEN

ABSTRACT: SETBP1 mutations are found in various clonal myeloid disorders. However, it is unclear whether they can initiate leukemia, because SETBP1 mutations typically appear as later events during oncogenesis. To answer this question, we generated a mouse model expressing mutated SETBP1 in hematopoietic tissue: this model showed profound alterations in the differentiation program of hematopoietic progenitors and developed a myeloid neoplasm with megakaryocytic dysplasia, splenomegaly, and bone marrow fibrosis, prompting us to investigate SETBP1 mutations in a cohort of 36 triple-negative primary myelofibrosis (TN-PMF) cases. We identified 2 distinct subgroups, one carrying SETBP1 mutations and the other completely devoid of somatic variants. Clinically, a striking difference in disease aggressiveness was noted, with patients with SETBP1 mutation showing a much worse clinical course. In contrast to myelodysplastic/myeloproliferative neoplasms, in which SETBP1 mutations are mostly found as a late clonal event, single-cell clonal hierarchy reconstruction in 3 patients with TN-PMF from our cohort revealed SETBP1 to be a very early event, suggesting that the phenotype of the different SETBP1+ disorders may be shaped by the opposite hierarchy of the same clonal SETBP1 variants.


Asunto(s)
Sistema Hematopoyético , Enfermedades Mielodisplásicas-Mieloproliferativas , Trastornos Mieloproliferativos , Mielofibrosis Primaria , Animales , Ratones , Humanos , Mielofibrosis Primaria/genética , Trastornos Mieloproliferativos/genética , Mutación , Proteínas Portadoras/genética , Proteínas Nucleares/genética
2.
Dev Biol ; 434(2): 231-248, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29305158

RESUMEN

During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Corteza Cerebral/embriología , Proteínas de Unión al ADN/metabolismo , Células-Madre Neurales/metabolismo , Organogénesis/efectos de los fármacos , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Corteza Cerebral/citología , Proteínas de Unión al ADN/genética , Ratones , Células-Madre Neurales/citología , Organogénesis/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética
3.
Mol Autism ; 14(1): 20, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37264456

RESUMEN

BACKGROUND: Neurodevelopmental disorders (NDDs) are heterogeneous conditions due to alterations of a variety of molecular mechanisms and cell dysfunctions. SETD5 haploinsufficiency leads to NDDs due to chromatin defects. Epigenetic basis of NDDs has been reported in an increasing number of cases while mitochondrial dysfunctions are more common within NDD patients than in the general population. METHODS: We investigated in vitro neural stem cells as well as the brain of the Setd5 haploinsufficiency mouse model interrogating its transcriptome, analyzing mitochondrial structure, biochemical composition, and dynamics, as well as mitochondrial functionality. RESULTS: Mitochondrial impairment is facilitated by transcriptional aberrations originated by the decrease of the SETD5 enzyme. Low levels of SETD5 resulted in fragmented mitochondria, reduced mitochondrial membrane potential, and ATP production both in neural precursors and neurons. Mitochondria were also mislocalized in mutant neurons, with reduced organelles within neurites and synapses. LIMITATIONS: We found several defects in the mitochondrial compartment; however, we can only speculate about their position in the hierarchy of the pathological mechanisms at the basis of the disease. CONCLUSIONS: Our study explores the interplay between chromatin regulation and mitochondria functions as a possible important aspect of SETD5-associated NDD pathophysiology. Our data, if confirmed in patient context, suggest that the mitochondrial activity and dynamics may represent new therapeutic targets for disorders associated with the loss of SETD5.


Asunto(s)
Haploinsuficiencia , Células-Madre Neurales , Ratones , Animales , Humanos , Neuronas/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/metabolismo , Cromatina/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo
4.
Nat Commun ; 14(1): 3212, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270547

RESUMEN

Within the chromatin, distal elements interact with promoters to regulate specific transcriptional programs. Histone acetylation, interfering with the net charges of the nucleosomes, is a key player in this regulation. Here, we report that the oncoprotein SET is a critical determinant for the levels of histone acetylation within enhancers. We disclose that a condition in which SET is accumulated, the severe Schinzel-Giedion Syndrome (SGS), is characterized by a failure in the usage of the distal regulatory regions typically employed during fate commitment. This is accompanied by the usage of alternative enhancers leading to a massive rewiring of the distal control of the gene transcription. This represents a (mal)adaptive mechanism that, on one side, allows to achieve a certain degree of differentiation, while on the other affects the fine and corrected maturation of the cells. Thus, we propose the differential in cis-regulation as a contributing factor to the pathological basis of SGS and possibly other the SET-related disorders in humans.


Asunto(s)
Elementos de Facilitación Genéticos , Histonas , Humanos , Histonas/genética , Histonas/metabolismo , Elementos de Facilitación Genéticos/genética , Diferenciación Celular/genética , Cromatina/genética , Regiones Promotoras Genéticas/genética
5.
Sci Adv ; 8(31): eabn3986, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35921410

RESUMEN

Current therapies remain unsatisfactory in preventing the recurrence of glioblastoma multiforme (GBM), which leads to poor patient survival. By rational engineering of the transcription factor SOX2, a key promoter of GBM malignancy, together with the Kruppel-associated box and DNA methyltransferase3A/L catalytic domains, we generated a synthetic repressor named SOX2 epigenetic silencer (SES), which induces the transcriptional silencing of its original targets. By doing so, SES kills both glioma cell lines and patient-derived cancer stem cells in vitro and in vivo. SES expression, through local viral delivery in mouse xenografts, induces strong regression of human tumors and survival rescue. Conversely, SES is not harmful to neurons and glia, also thanks to a minimal promoter that restricts its expression in mitotically active cells, rarely present in the brain parenchyma. Collectively, SES produces a significant silencing of a large fraction of the SOX2 transcriptional network, achieving high levels of efficacy in repressing aggressive brain tumors.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Epigénesis Genética , Glioblastoma/metabolismo , Glioma/patología , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
6.
Biomolecules ; 11(11)2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34827709

RESUMEN

The advent of next-generation sequencing (NGS) is heavily changing both the diagnosis of human conditions and basic biological research. It is now possible to dig deep inside the genome of hundreds of thousands or even millions of people and find both common and rare genomic variants and to perform detailed phenotypic characterizations of both physiological organs and experimental models. Recent years have seen the introduction of multiple techniques using NGS to profile transcription, DNA and chromatin modifications, protein binding, etc., that are now allowing us to profile cells in bulk or even at a single-cell level. Although rare and ultra-rare diseases only affect a few people, each of these diseases represent scholarly cases from which a great deal can be learned about the pathological and physiological function of genes, pathways, and mechanisms. Therefore, for rare diseases, state-of-the-art investigations using NGS have double valence: their genomic cause (new variants) and the characterize the underlining the mechanisms associated with them (discovery of gene function) can be found. In a non-exhaustive manner, this review will outline the main usage of NGS-based techniques for the diagnosis and characterization of neurodevelopmental disorders (NDDs), under whose umbrella many rare and ultra-rare diseases fall.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedades Raras , Humanos , Análisis de Secuencia de ADN
7.
Front Cell Dev Biol ; 9: 641410, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708771

RESUMEN

TBL1XR1 gene is associated with multiple developmental disorders presenting several neurological aspects. The relative protein is involved in the modulation of important cellular pathways and master regulators of transcriptional output, including nuclear receptor repressors, Wnt signaling, and MECP2 protein. However, TBL1XR1 mutations (including complete loss of its functions) have not been experimentally studied in a neurological context, leaving a knowledge gap in the mechanisms at the basis of the diseases. Here, we show that Tbl1xr1 knock-out mice exhibit behavioral and neuronal abnormalities. Either the absence of TBL1XR1 or its point mutations interfering with stability/regulation of NCOR complex induced decreased proliferation and increased differentiation in neural progenitors. We suggest that this developmental unbalance is due to a failure in the regulation of the MAPK cascade. Taken together, our results broaden the molecular and functional aftermath of TBL1XR1 deficiency associated with human disorders.

8.
Nat Commun ; 12(1): 4050, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193871

RESUMEN

The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.


Asunto(s)
Anomalías Múltiples/patología , Proteínas Portadoras/metabolismo , Anomalías Craneofaciales/patología , Daño del ADN , Deformidades Congénitas de la Mano/patología , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Discapacidad Intelectual/patología , Mutación , Uñas Malformadas/patología , Células-Madre Neurales/patología , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/metabolismo , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Uñas Malformadas/genética , Uñas Malformadas/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , Organoides
9.
Nat Commun ; 11(1): 4178, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826895

RESUMEN

Friedreich's ataxia (FRDA) is an autosomal-recessive neurodegenerative and cardiac disorder which occurs when transcription of the FXN gene is silenced due to an excessive expansion of GAA repeats into its first intron. Herein, we generate dorsal root ganglia organoids (DRG organoids) by in vitro differentiation of human iPSCs. Bulk and single-cell RNA sequencing show that DRG organoids present a transcriptional signature similar to native DRGs and display the main peripheral sensory neuronal and glial cell subtypes. Furthermore, when co-cultured with human intrafusal muscle fibers, DRG organoid sensory neurons contact their peripheral targets and reconstitute the muscle spindle proprioceptive receptors. FRDA DRG organoids model some molecular and cellular deficits of the disease that are rescued when the entire FXN intron 1 is removed, and not with the excision of the expanded GAA tract. These results strongly suggest that removal of the repressed chromatin flanking the GAA tract might contribute to rescue FXN total expression and fully revert the pathological hallmarks of FRDA DRG neurons.


Asunto(s)
Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Ganglios Espinales/metabolismo , Edición Génica/métodos , Proteínas de Unión a Hierro/genética , Organoides/metabolismo , Células Receptoras Sensoriales/metabolismo , Antioxidantes/farmacología , Sistemas CRISPR-Cas , Diferenciación Celular , Cromatina/metabolismo , Ataxia de Friedreich/tratamiento farmacológico , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Predisposición Genética a la Enfermedad/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Intrones , Mitocondrias/metabolismo , Organoides/efectos de los fármacos , Organoides/patología , Células Receptoras Sensoriales/patología , Análisis de Secuencia de ARN , Transcriptoma , Frataxina
10.
Front Genet ; 10: 1291, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31998360

RESUMEN

Post-translational methylation of H3 lysine 36 (H3K36) is an important epigenetic marker that majorly contributes to the functionality of the chromatin. This mark is interpreted by the cell in several crucial biological processes including gene transcription and DNA methylation. The homeostasis of H3K36 methylation is finely regulated by different enzyme classes which, when impaired, lead to a plethora of diseases; ranging from multi-organ syndromes to cancer, to pure neurological diseases often associated with brain development. This mini-review summarizes current knowledge on these important epigenetic signals with emphasis on the molecular mechanisms that (i) regulate their abundance, (ii) are influenced by H3K36 methylation, and (iii) the associated diseases.

11.
Neuron ; 104(2): 271-289.e13, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31515109

RESUMEN

Mutations in one SETD5 allele are genetic causes of intellectual disability and autistic spectrum disorders. However, the mechanisms by which SETD5 regulates brain development and function remain largely elusive. Herein, we found that Setd5 haploinsufficiency impairs the proliferative dynamics of neural progenitors and synaptic wiring of neurons, ultimately resulting in behavioral deficits in mice. Mechanistically, Setd5 inactivation in neural stem cells, zebrafish, and mice equally affects genome-wide levels of H3K36me3 on active gene bodies. Notably, we demonstrated that SETD5 directly deposits H3K36me3, which is essential to allow on-time RNA elongation dynamics. Hence, Setd5 gene loss leads to abnormal transcription, with impaired RNA maturation causing detrimental effects on gene integrity and splicing. These findings identify SETD5 as a fundamental epigenetic enzyme controlling the transcriptional landscape in neural progenitors and their derivatives and illuminate the molecular events that connect epigenetic defects with neuronal dysfunctions at the basis of related human diseases.


Asunto(s)
Encéfalo/embriología , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Código de Histonas/genética , Metiltransferasas/genética , Proteínas de Pez Cebra/fisiología , Animales , Conducta Animal , Encéfalo/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Cognición , Epigénesis Genética , Histona Metiltransferasas/genética , Metiltransferasas/fisiología , Ratones , Mutación , Células-Madre Neurales/metabolismo , Empalme del ARN/genética , RNA-Seq , Conducta Social , Elongación de la Transcripción Genética , Pez Cebra , Proteínas de Pez Cebra/genética
12.
J Mol Neurosci ; 61(4): 590-602, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28255957

RESUMEN

Epigenetics is the array of the chromatin modifications that customize in cell-, stage-, or condition-specific manner the information encloses in plain DNA molecules. Increasing evidences suggest the importance of epigenetic mechanisms for development and maintenance of central nervous system. In fact, a large number of newly discovered genetic causes of neurodevelopmental disorders such as intellectual disability, autism spectrum disorders, and many other syndromes are mutations within genes encoding for chromatin remodeling enzymes. Here, we review recent findings on the epigenetic origin of human diseases, with emphasis on disorders that affect development of the nervous system, and discuss novel therapeutic avenues that target epigenetic mechanisms.


Asunto(s)
Epigénesis Genética , Trastornos del Neurodesarrollo/genética , Animales , Ensamble y Desensamble de Cromatina , Metilación de ADN , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA