Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 30: 465-502, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25000993

RESUMEN

Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.


Asunto(s)
Neocórtex/crecimiento & desarrollo , Neurogénesis/fisiología , Animales , División Celular Asimétrica , Compartimento Celular , Linaje de la Célula , Membrana Celular/fisiología , Núcleo Celular/fisiología , Polaridad Celular , Líquido Cefalorraquídeo/fisiología , Humanos , Uniones Intercelulares/fisiología , Ventrículos Laterales/embriología , Lípidos de la Membrana/metabolismo , Microglía/fisiología , Mitosis , Neocórtex/citología , Neocórtex/embriología , Células-Madre Neurales/clasificación , Células-Madre Neurales/fisiología , Células Neuroepiteliales/citología , Células Neuroepiteliales/fisiología , Neuronas/fisiología , Orgánulos/fisiología , Especificidad de la Especie
2.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37665322

RESUMEN

One-carbon/folate (1C) metabolism supplies methyl groups required for DNA and histone methylation, and is involved in the maintenance of self-renewal in stem cells. Dihydrofolate reductase (DHFR), a key enzyme in 1C metabolism, is highly expressed in human and mouse neural progenitors at the early stages of neocortical development. Here, we have investigated the role of DHFR in the developing neocortex and report that reducing its activity in human neural organoids and mouse embryonic neocortex accelerates indirect neurogenesis, thereby affecting neuronal composition of the neocortex. Furthermore, we show that decreasing DHFR activity in neural progenitors leads to a reduction in one-carbon/folate metabolites and correlates with modifications of H3K4me3 levels. Our findings reveal an unanticipated role for DHFR in controlling specific steps of neocortex development and indicate that variations in 1C metabolic cues impact cell fate transitions.


Asunto(s)
Neocórtex , Neurogénesis , Tetrahidrofolato Deshidrogenasa , Animales , Humanos , Ratones , Carbono , Ácido Fólico , Neurogénesis/genética , Tetrahidrofolato Deshidrogenasa/genética
3.
Genes Dev ; 32(2): 165-180, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440260

RESUMEN

Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.


Asunto(s)
ADN Helicasas/fisiología , Proteínas de Unión al ADN/fisiología , Epigénesis Genética , Células-Madre Neurales/metabolismo , Células Neuroepiteliales/metabolismo , Síndrome CHARGE/genética , Línea Celular , Linaje de la Célula/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Haploinsuficiencia , Humanos , Cresta Neural/metabolismo , Transcripción Genética
4.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199563

RESUMEN

In contrast to stage-specific transcription factors, the role of ubiquitous transcription factors in neuronal development remains a matter of scrutiny. Here, we demonstrated that a ubiquitous factor NF-Y is essential for neural progenitor maintenance during brain morphogenesis. Deletion of the NF-YA subunit in neural progenitors by using nestin-cre transgene in mice resulted in significant abnormalities in brain morphology, including a thinner cerebral cortex and loss of striatum during embryogenesis. Detailed analyses revealed a progressive decline in multiple neural progenitors in the cerebral cortex and ganglionic eminences, accompanied by induced apoptotic cell death and reduced cell proliferation. In neural progenitors, the NF-YA short isoform lacking exon 3 is dominant and co-expressed with cell cycle genes. ChIP-seq analysis from the cortex during early corticogenesis revealed preferential binding of NF-Y to the cell cycle genes, some of which were confirmed to be downregulated following NF-YA deletion. Notably, the NF-YA short isoform disappears and is replaced by its long isoform during neuronal differentiation. Forced expression of the NF-YA long isoform in neural progenitors resulted in a significant decline in neuronal count, possibly due to the suppression of cell proliferation. Collectively, we elucidated a critical role of the NF-YA short isoform in maintaining neural progenitors, possibly by regulating cell proliferation and apoptosis. Moreover, we identified an isoform switch in NF-YA within the neuronal lineage in vivo, which may explain the stage-specific role of NF-Y during neuronal development.


Asunto(s)
Factor de Unión a CCAAT , Corteza Cerebral , Animales , Ratones , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica , Neurogénesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo
5.
Mol Cell ; 65(3): 373-375, 2017 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-28157500

RESUMEN

Polycomb proteins are well-known epigenetic repressors with unexplained roles in chromatin folding. In this issue of Molecular Cell, Kundu et al. (2017) investigate the structures of PRC1-mediated domains in stem cells and probe their changes upon differentiation and in PRC knockouts.


Asunto(s)
Células Madre Embrionarias/citología , Complejo Represivo Polycomb 1/química , Complejo Represivo Polycomb 1/metabolismo , Animales , Diferenciación Celular , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Humanos , Complejo Represivo Polycomb 1/genética , Dominios Proteicos
6.
Bioessays ; 45(9): e2300039, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37439444

RESUMEN

Heterogeneous nuclear ribonucleoprotein U (HNRNPU) is a nuclear protein that plays a crucial role in various biological functions, such as RNA splicing and chromatin organization. HNRNPU/scaffold attachment factor A (SAF-A) activities are essential for regulating gene expression, DNA replication, genome integrity, and mitotic fidelity. These functions are critical to ensure the robustness of developmental processes, particularly those involved in shaping the human brain. As a result, HNRNPU is associated with various neurodevelopmental disorders (HNRNPU-related neurodevelopmental disorder, HNRNPU-NDD) characterized by developmental delay and intellectual disability. Our research demonstrates that the loss of HNRNPU function results in the death of both neural progenitor cells and post-mitotic neurons, with a higher sensitivity observed in the former. We reported that HNRNPU truncation leads to the dysregulation of gene expression and alternative splicing of genes that converge on several signaling pathways, some of which are likely to be involved in the pathology of HNRNPU-related NDD.


Asunto(s)
Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo U/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Encéfalo/metabolismo , Genoma , Empalme del ARN
7.
J Cell Physiol ; 239(5): e31249, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38501376

RESUMEN

The hippocampal dentate gyrus, responds to diverse pathological stimuli through neurogenesis. This phenomenon, observed following brain injury or neurodegeneration, is postulated to contribute to neuronal repair and functional recovery, thereby presenting an avenue for endogenous neuronal restoration. This study investigated the extent of regenerative response in hippocampal neurogenesis by leveraging the well-established kainic acid-induced status epilepticus model in vivo. In our study, we observed the activation and proliferation of neuronal progenitors or neural stem cell (NSC) and their subsequent migration to the injury sites following the seizure. At the injury sites, new neurons (Tuj1+BrdU+ and NeuN+BrdU+) have been generated indicating regenerative and reparative roles of the progenitor cells. We further detected whether this transient neurogenic burst, which might be a response towards an attempt to repair the brain, is associated with persistent long-term exhaustion of the dentate progenitor cells and impairment of adult neurogenesis marked by downregulation of Ki67, HoPX, and Sox2 with BrdU+ cell in the later part of life. Our studies suggest that the adult brain has the constitutive endogenous regenerative potential for brain repair to restore the damaged neurons, meanwhile, in the long term, it accelerates the depletion of the finite NSC pool in the hippocampal neurogenic niche by changing its proliferative and neurogenic capacity. A thorough understanding of the impact of modulating adult neurogenesis will eventually be required to design novel therapeutics to stimulate or assist brain repair while simultaneously preventing the adverse effects of early robust neurogenesis on the proliferative potential of endogenous neuronal progenitors.


Asunto(s)
Hipocampo , Células-Madre Neurales , Neurogénesis , Animales , Células-Madre Neurales/metabolismo , Hipocampo/patología , Hipocampo/metabolismo , Proliferación Celular , Masculino , Nicho de Células Madre , Giro Dentado/patología , Giro Dentado/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Ácido Kaínico/toxicidad , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología , Estado Epiléptico/metabolismo , Regeneración Nerviosa , Modelos Animales de Enfermedad , Ratones , Movimiento Celular
8.
Neurobiol Dis ; 199: 106550, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38849103

RESUMEN

Bioenergetics describe the biochemical processes responsible for energy supply in organisms. When these changes become dysregulated in brain development, multiple neurodevelopmental diseases can occur, implicating bioenergetics as key regulators of neural development. Historically, the discovery of disease processes affecting individual stages of brain development has revealed critical roles that bioenergetics play in generating the nervous system. Bioenergetic-dependent neurodevelopmental disorders include neural tube closure defects, microcephaly, intellectual disability, autism spectrum disorders, epilepsy, mTORopathies, and oncogenic processes. Developmental timing and cell-type specificity of these changes determine the long-term effects of bioenergetic disease mechanisms on brain form and function. Here, we discuss key metabolic regulators of neural progenitor specification, neuronal differentiation (neurogenesis), and gliogenesis. In general, transitions between glycolysis and oxidative phosphorylation are regulated in early brain development and in oncogenesis, and reactive oxygen species (ROS) and mitochondrial maturity play key roles later in differentiation. We also discuss how bioenergetics interface with the developmental regulation of other key neural elements, including the cerebrospinal fluid brain environment. While questions remain about the interplay between bioenergetics and brain development, this review integrates the current state of known key intersections between these processes in health and disease.


Asunto(s)
Encéfalo , Metabolismo Energético , Neurogénesis , Humanos , Metabolismo Energético/fisiología , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Animales , Neurogénesis/fisiología , Trastornos del Neurodesarrollo/metabolismo
9.
Genet Med ; 26(4): 101057, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38158856

RESUMEN

PURPOSE: We established the genetic etiology of a syndromic neurodevelopmental condition characterized by variable cognitive impairment, recognizable facial dysmorphism, and a constellation of extra-neurological manifestations. METHODS: We performed phenotypic characterization of 6 participants from 4 unrelated families presenting with a neurodevelopmental syndrome and used exome sequencing to investigate the underlying genetic cause. To probe relevance to the neurodevelopmental phenotype and craniofacial dysmorphism, we established two- and three-dimensional human stem cell-derived neural models and generated a stable cachd1 zebrafish mutant on a transgenic cartilage reporter line. RESULTS: Affected individuals showed mild cognitive impairment, dysmorphism featuring oculo-auriculo abnormalities, and developmental defects involving genitourinary and digestive tracts. Exome sequencing revealed biallelic putative loss-of-function variants in CACHD1 segregating with disease in all pedigrees. RNA sequencing in CACHD1-depleted neural progenitors revealed abnormal expression of genes with key roles in Wnt signaling, neurodevelopment, and organ morphogenesis. CACHD1 depletion in neural progenitors resulted in reduced percentages of post-mitotic neurons and enlargement of 3D neurospheres. Homozygous cachd1 mutant larvae showed mandibular patterning defects mimicking human facial dysmorphism. CONCLUSION: Our findings support the role of loss-of-function variants in CACHD1 as the cause of a rare neurodevelopmental syndrome with facial dysmorphism and multisystem abnormalities.


Asunto(s)
Anomalías Múltiples , Anomalías Craneofaciales , Anomalías Musculoesqueléticas , Trastornos del Neurodesarrollo , Animales , Humanos , Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Anomalías Musculoesqueléticas/genética , Trastornos del Neurodesarrollo/genética , Fenotipo , Síndrome , Pez Cebra/genética
10.
Vet Res ; 55(1): 32, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38493182

RESUMEN

Outbreaks of West Nile virus (WNV) occur periodically, affecting both human and equine populations. There are no vaccines for humans, and those commercialised for horses do not have sufficient coverage. Specific antiviral treatments do not exist. Many drug discovery studies have been conducted, but since rodent or primate cell lines are normally used, results cannot always be transposed to horses. There is thus a need to develop relevant equine cellular models. Here, we used induced pluripotent stem cells to develop a new in vitro model of WNV-infected equine brain cells suitable for microplate assay, and assessed the cytotoxicity and antiviral activity of forty-one chemical compounds. We found that one nucleoside analog, 2'C-methylcytidine, blocked WNV infection in equine brain cells, whereas other compounds were either toxic or ineffective, despite some displaying anti-viral activity in human cell lines. We also revealed an unexpected proviral effect of statins in WNV-infected equine brain cells. Our results thus identify a potential lead for future drug development and underscore the importance of using a tissue- and species-relevant cellular model for assessing the activity of antiviral compounds.


Asunto(s)
Enfermedades de los Caballos , Células Madre Pluripotentes Inducidas , Fiebre del Nilo Occidental , Virus del Nilo Occidental , Animales , Caballos , Humanos , Fiebre del Nilo Occidental/veterinaria , Fiebre del Nilo Occidental/epidemiología , Encéfalo , Antivirales/farmacología , Antivirales/uso terapéutico , Enfermedades de los Caballos/tratamiento farmacológico
11.
Cell Mol Life Sci ; 80(1): 36, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36627412

RESUMEN

Cell differentiation involves profound changes in global gene expression that often has to occur in coordination with cell cycle exit. Because cyclin-dependent kinase inhibitor p27 reportedly regulates proliferation of neural progenitor cells in the subependymal neurogenic niche of the adult mouse brain, but can also have effects on gene expression, we decided to molecularly analyze its role in adult neurogenesis and oligodendrogenesis. At the cell level, we show that p27 restricts residual cyclin-dependent kinase activity after mitogen withdrawal to antagonize cycling, but it is not essential for cell cycle exit. By integrating genome-wide gene expression and chromatin accessibility data, we find that p27 is coincidentally necessary to repress many genes involved in the transit from multipotentiality to differentiation, including those coding for neural progenitor transcription factors SOX2, OLIG2 and ASCL1. Our data reveal both a direct association of p27 with regulatory sequences in the three genes and an additional hierarchical relationship where p27 repression of Sox2 leads to reduced levels of its downstream targets Olig2 and Ascl1. In vivo, p27 is also required for the regulation of the proper level of SOX2 necessary for neuroblasts and oligodendroglial progenitor cells to timely exit cell cycle in a lineage-dependent manner.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Neurogénesis , Factores de Transcripción SOXB1 , Animales , Ratones , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , División Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Expresión Génica , Neurogénesis/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
12.
Int J Mol Sci ; 25(16)2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39201803

RESUMEN

The degeneration of spiral ganglion neurons (SGNs), which convey auditory signals from hair cells to the brain, can be a primary cause of sensorineural hearing loss (SNHL) or can occur secondary to hair cell loss. Emerging therapies for SNHL include the replacement of damaged SGNs using stem cell-derived otic neuronal progenitors (ONPs). However, the availability of renewable, accessible, and patient-matched sources of human stem cells is a prerequisite for successful replacement of the auditory nerve. In this study, we derived ONP and SGN-like cells by a reliable and reproducible stepwise guidance differentiation procedure of self-renewing human dental pulp stem cells (hDPSCs). This in vitro differentiation protocol relies on the modulation of BMP and TGFß pathways using a free-floating 3D neurosphere method, followed by differentiation on a Geltrex-coated surface using two culture paradigms to modulate the major factors and pathways involved in early otic neurogenesis. Gene and protein expression analyses revealed efficient induction of a comprehensive panel of known ONP and SGN-like cell markers during the time course of hDPSCs differentiation. Atomic force microscopy revealed that hDPSC-derived SGN-like cells exhibit similar nanomechanical properties as their in vivo SGN counterparts. Furthermore, spiral ganglion neurons from newborn rats come in close contact with hDPSC-derived ONPs 5 days after co-culturing. Our data demonstrate the capability of hDPSCs to generate SGN-like neurons with specific lineage marker expression, bipolar morphology, and the nanomechanical characteristics of SGNs, suggesting that the neurons could be used for next-generation cochlear implants and/or inner ear cell-based strategies for SNHL.


Asunto(s)
Diferenciación Celular , Pulpa Dental , Neuronas , Ganglio Espiral de la Cóclea , Pulpa Dental/citología , Humanos , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/metabolismo , Animales , Ratas , Neuronas/metabolismo , Neuronas/citología , Células Cultivadas , Nervio Coclear/citología , Nervio Coclear/metabolismo , Células Madre/citología , Células Madre/metabolismo , Neurogénesis
13.
Dev Dyn ; 252(3): 363-376, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36153792

RESUMEN

BACKGROUND: The apical surface (AS) of epithelial cells is highly specialized; it is important for morphogenetic processes that are essential to shape organs and tissues and it plays a role in morphogen and growth factor signaling. Apical progenitors in the mammalian neocortex are pseudoepithelial cells whose apical surface lines the ventricle. Whether changes in their apical surface sizes are important for cortical morphogenesis and/or other aspects of neocortex development has not been thoroughly addressed. RESULTS: Here we show that apical progenitors are heterogeneous with respect to their apical surface area. In Efnb1 mutants, the size of the apical surface is modified and this correlates with discrete alterations of tissue organization without impacting apical progenitors proliferation. CONCLUSIONS: Altogether, our data reveal heterogeneity in apical progenitors AS area in the developing neocortex and shows a role for Ephrin B1 in controlling AS size. Our study also indicates that changes in AS size do not have strong repercussion on apical progenitor behavior.


Asunto(s)
Neocórtex , Neuronas , Animales , Neuronas/metabolismo , Transducción de Señal , Efrina-B1/metabolismo , Mamíferos/metabolismo
14.
J Neurosci ; 42(3): 362-376, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34819341

RESUMEN

Multifaceted microglial functions in the developing brain, such as promoting the differentiation of neural progenitors and contributing to the positioning and survival of neurons, have been progressively revealed. Although previous studies have noted the relationship between vascular endothelial cells and microglia in the developing brain, little attention has been given to the importance of pericytes, the mural cells surrounding endothelial cells. In this study, we attempted to dissect the role of pericytes in microglial distribution and function in developing mouse brains. Our immunohistochemical analysis showed that approximately half of the microglia attached to capillaries in the cerebral walls. Notably, a magnified observation of the position of microglia, vascular endothelial cells and pericytes demonstrated that microglia were preferentially associated with pericytes that covered 79.8% of the total capillary surface area. Through in vivo pericyte depletion induced by the intraventricular administration of a neutralizing antibody against platelet-derived growth factor receptor (PDGFR)ß (clone APB5), we found that microglial density was markedly decreased compared with that in control antibody-treated brains because of their low proliferative capacity. Moreover, in vitro coculture of isolated CD11b+ microglia and NG2+PDGFRα- cells, which are mostly composed of pericytes, from parenchymal cells indicated that pericytes promote microglial proliferation via the production of soluble factors. Furthermore, pericyte depletion by APB5 treatment resulted in a failure of microglia to promote the differentiation of neural stem cells into intermediate progenitors. Taken together, our findings suggest that pericytes facilitate microglial homeostasis in the developing brains, thereby indirectly supporting microglial effects on neural progenitors.SIGNIFICANCE STATEMENT This study highlights the novel effect of pericytes on microglia in the developing mouse brain. Through multiple analyses using an in vivo pericyte depletion mouse model and an in vitro coculture study of isolated pericytes and microglia from parenchymal cells, we demonstrated that pericytes contribute to microglial proliferation and support microglia in efficiently promoting the differentiation of neural stem cells into intermediate progenitors. Our present data provide evidence that pericytes function not only in the maintenance of cerebral microcirculation and blood brain barrier (BBB) integrity but also in microglial homeostasis in the developing cerebral walls. These findings will expand our knowledge and help elucidate the mechanism of brain development both in healthy and disease conditions.


Asunto(s)
Corteza Cerebral/citología , Homeostasis/fisiología , Microglía/citología , Células-Madre Neurales/citología , Pericitos/citología , Animales , Anticuerpos Neutralizantes , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/embriología , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Ácido Clodrónico/farmacología , Homeostasis/efectos de los fármacos , Liposomas , Ratones , Microglía/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Pericitos/efectos de los fármacos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas
15.
Dev Biol ; 481: 116-128, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34666024

RESUMEN

During development, neural progenitors undergo temporal patterning as they age to sequentially generate differently fated progeny. Temporal patterning of neural progenitors is relatively well-studied in Drosophila. Temporal cascades of transcription factors or opposing temporal gradients of RNA-binding proteins are expressed in neural progenitors as they age to control the fates of the progeny. The temporal progression is mostly driven by intrinsic mechanisms including cross-regulations between temporal genes, but environmental cues also play important roles in certain transitions. Vertebrate neural progenitors demonstrate greater plasticity in response to extrinsic cues. Recent studies suggest that vertebrate neural progenitors are also temporally patterned by a combination of transcriptional and post-transcriptional mechanisms in response to extracellular signaling to regulate neural fate specification. In this review, we summarize recent advances in the study of temporal patterning of neural progenitors in Drosophila and vertebrates. We also discuss the involvement of epigenetic mechanisms, specifically the Polycomb group complexes and ATP-dependent chromatin remodeling complexes, in the temporal patterning of neural progenitors.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Neurogénesis , Transducción de Señal , Animales , Drosophila melanogaster
16.
Development ; 147(22)2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33060132

RESUMEN

The mammalian cortex is populated by neurons derived from neural progenitors located throughout the embryonic telencephalon. Excitatory neurons are derived from the dorsal telencephalon, whereas inhibitory interneurons are generated in its ventral portion. The transcriptional regulator PRDM16 is expressed by radial glia, neural progenitors present in both regions; however, its mechanisms of action are still not fully understood. It is unclear whether PRDM16 plays a similar role in neurogenesis in both dorsal and ventral progenitor lineages and, if so, whether it regulates common or unique networks of genes. Here, we show that Prdm16 expression in mouse medial ganglionic eminence (MGE) progenitors is required for maintaining their proliferative capacity and for the production of proper numbers of forebrain GABAergic interneurons. PRDM16 binds to cis-regulatory elements and represses the expression of region-specific neuronal differentiation genes, thereby controlling the timing of neuronal maturation. PRDM16 regulates convergent developmental gene expression programs in the cortex and MGE, which utilize both common and region-specific sets of genes to control the proliferative capacity of neural progenitors, ensuring the generation of correct numbers of cortical neurons.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción/metabolismo , Animales , Corteza Cerebral/citología , Proteínas de Unión al ADN/genética , Neuronas GABAérgicas/citología , Interneuronas/citología , Ratones , Células-Madre Neurales/citología , Factores de Transcripción/genética
17.
Stem Cells ; 40(7): 678-690, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35429390

RESUMEN

RNF113A (Ring Finger Protein 113A) is genetically associated with autism spectrum disorders and X-linked trichothiodystrophy (TTD) syndrome. Loss-of-function mutations in human RNF113A are causally linked to TTD, which is characterized by abnormal development of the central nervous system (CNS) and mental retardation. How the loss of RNF113A activity affects brain development is not known. Here we identify Rnf113a1 as a critical regulator of cell death and neurogenesis during mouse brain development. Rnf113a1 gene exhibits widespread expression in the embryonic CNS. Knockdown studies in embryonic cortical neural stem/progenitor cells (NSCs) and the mouse cortex suggest that Rnf113a1 controls the survival, proliferation, and differentiation properties of progenitor cells. Importantly, Rnf113a1 deficiency triggers cell apoptosis via a combined action on essential regulators of cell survival, including p53, Nupr1, and Rad51. Collectively, these observations establish Rnf113a1 as a regulatory factor in CNS development and provide insights into its role in neurodevelopmental defects associated with TTD and autism.


Asunto(s)
Células-Madre Neurales , Trastornos del Neurodesarrollo , Animales , Apoptosis/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Células-Madre Neurales/metabolismo , Trastornos del Neurodesarrollo/genética , Neurogénesis/fisiología
18.
Stem Cells ; 40(2): 175-189, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35257173

RESUMEN

Hox genes play key roles in the anterior-posterior (AP) specification of all 3 germ layers during different developmental stages. It is only partially understood how they function in widely different developmental contexts, particularly with regards to extracellular signaling, and to what extent their function can be harnessed to guide cell specification in vitro. Here, we addressed the role of Hoxb1 in 2 distinct developmental contexts; in mouse embryonic stem cells (mES)-derived neuromesodermal progenitors (NMPs) and hindbrain neural progenitors. We found that Hoxb1 promotes NMP survival through the upregulation of Fgf8, Fgf17, and other components of Fgf signaling as well as the repression of components of the apoptotic pathway. Additionally, it upregulates other anterior Hox genes suggesting that it plays an active role in the early steps of AP specification. In neural progenitors, Hoxb1 synergizes with shh to repress anterior and dorsal neural markers, promote the expression of ventral neural markers and direct the specification of facial branchiomotorneuron (FBM)-like progenitors. Hoxb1 and shh synergize in regulating the expression of diverse signals and signaling molecules, including the Ret tyrosine kinase receptor. Finally, Hoxb1 synergizes with exogenous Glial cell line-derived neurotrophic factor (GDNF) to strengthen Ret expression and further promote the generation of FBM-like progenitors. Facial branchiomotorneuron-like progenitors survived for at least 6 months and differentiated into postmitotic neurons after orthotopic transplantation near the facial nucleus of adult mice. These results suggested that the patterning activity of Hox genes in combination with downstream signaling molecules can be harnessed for the generation of defined neural populations and transplantations with implications for neurodegenerative diseases.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Rombencéfalo , Animales , Diferenciación Celular/genética , Supervivencia Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Rombencéfalo/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
19.
Proc Natl Acad Sci U S A ; 117(36): 22331-22340, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32839322

RESUMEN

The chromatin remodeler CHD8 is among the most frequently mutated genes in autism spectrum disorder (ASD). CHD8 has a dosage-sensitive role in ASD, but when and how it becomes critical to human social function is unclear. Here, we conducted genomic analyses of heterozygous and homozygous Chd8 mouse embryonic stem cells and differentiated neural progenitors. We identify dosage-sensitive CHD8 transcriptional targets, sites of regulated accessibility, and an unexpected cooperation with SOX transcription factors. Collectively, our findings reveal that CHD8 negatively regulates expression of neuronal genes to maintain pluripotency and also during differentiation. Thus, CHD8 is essential for both the maintenance of pluripotency and neural differentiation, providing mechanistic insight into its function with potential implications for ASD.


Asunto(s)
Proteínas de Unión al ADN , Dosificación de Gen/genética , Neurogénesis/genética , Animales , Trastorno del Espectro Autista , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Ratones , Ratones Noqueados
20.
Genes Dev ; 29(23): 2504-15, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26584621

RESUMEN

During spinal cord development, ventral neural progenitor cells that express the transcription factors Olig1 and Olig2, called pMN progenitors, produce motor neurons and then oligodendrocytes. Whether motor neurons and oligodendrocytes arise from common or distinct progenitors in vivo is not known. Using zebrafish, we found that motor neurons and oligodendrocytes are produced sequentially by distinct progenitors that have distinct origins. When olig2(+) cells were tracked during the peak period of motor neuron formation, most differentiated as motor neurons without further cell division. Using time-lapse imaging, we found that, as motor neurons differentiated, more dorsally positioned neuroepithelial progenitors descended to the pMN domain and initiated olig2 expression. Inhibition of Hedgehog signaling during motor neuron differentiation blocked the ventral movement of progenitors, the progressive initiation of olig2 expression, and oligodendrocyte formation. We therefore propose that the motor neuron-to-oligodendrocyte switch results from Hedgehog-mediated recruitment of glial-fated progenitors to the pMN domain subsequent to neurogenesis.


Asunto(s)
Diferenciación Celular , Neuronas Motoras/citología , Oligodendroglía/citología , Células Madre/citología , Pez Cebra/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula , Movimiento Celular , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células Neuroepiteliales/citología , Neurogénesis/fisiología , Factor de Transcripción 2 de los Oligodendrocitos , Estructura Terciaria de Proteína , Transducción de Señal , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA