Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 136(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37921122

RESUMEN

The covalent modification of histones is critical for many biological functions in mammals, including gene regulation and chromatin structure. Posttranslational histone modifications are added and removed by specialised 'writer' and 'eraser' enzymes, respectively. One such writer protein implicated in a wide range of cellular processes is SET domain-containing 2 (SETD2), a histone methyltransferase that catalyses the trimethylation of lysine 36 on histone H3 (H3K36me3). Recently, SETD2 has also been found to modify proteins other than histones, including actin and tubulin. The emerging roles of SETD2 in the development and function of the mammalian central nervous system (CNS) are of particular interest as several SETD2 variants have been implicated in neurodevelopmental disorders, such as autism spectrum disorder and the overgrowth disorder Luscan-Lumish syndrome. Here, we summarise the numerous roles of SETD2 in mammalian cellular functions and development, with a focus on the CNS. We also provide an overview of the consequences of SETD2 variants in human disease and discuss future directions for understanding essential cellular functions of SETD2.


Asunto(s)
Trastorno del Espectro Autista , Histonas , Animales , Humanos , Histonas/metabolismo , Trastorno del Espectro Autista/genética , Metilación , Cromatina , Sistema Nervioso Central/metabolismo , Mamíferos/metabolismo
2.
Development ; 149(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35245348

RESUMEN

The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape.


Asunto(s)
Desarrollo Embrionario/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Animales , Proliferación Celular/genética , Represión Epigenética/genética , Femenino , Masculino , Ratones , Mutación/genética , Transcriptoma/genética
3.
Cereb Cortex ; 34(7)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38960704

RESUMEN

The Polycomb Repressive Complex 2 (PRC2) regulates corticogenesis, yet the consequences of mutations to this epigenetic modifier in the mature brain are poorly defined. Importantly, PRC2 core genes are haploinsufficient and causative of several human neurodevelopmental disorders. To address the role of PRC2 in mature cortical structure and function, we conditionally deleted the PRC2 gene Eed from the developing mouse dorsal telencephalon. Adult homozygotes displayed smaller forebrain structures. Single-nucleus transcriptomics revealed that glutamatergic neurons were particularly affected, exhibiting dysregulated gene expression profiles, accompanied by aberrations in neuronal morphology and connectivity. Remarkably, homozygous mice performed well on challenging cognitive tasks. In contrast, while heterozygous mice did not exhibit clear anatomical or behavioral differences, they displayed dysregulation of neuronal genes and altered neuronal morphology that was strikingly different from homozygous phenotypes. Collectively, these data reveal how alterations to PRC2 function shape the mature brain and reveal a dose-specific role for PRC2 in determining glutamatergic neuron identity.


Asunto(s)
Ácido Glutámico , Neurogénesis , Neuronas , Complejo Represivo Polycomb 2 , Animales , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Ratones , Neurogénesis/fisiología , Ácido Glutámico/metabolismo , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Masculino , Ratones Endogámicos C57BL , Femenino , Ratones Transgénicos
4.
Development ; 148(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128986

RESUMEN

The balance between stem cell potency and lineage specification entails the integration of both extrinsic and intrinsic cues, which ultimately influence gene expression through the activity of transcription factors. One example of this is provided by the Hippo signalling pathway, which plays a central role in regulating organ size during development. Hippo pathway activity is mediated by the transcriptional co-factors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), which interact with TEA domain (TEAD) proteins to regulate gene expression. Although the roles of YAP and TAZ have been intensively studied, the roles played by TEAD proteins are less well understood. Recent studies have begun to address this, revealing that TEADs regulate the balance between progenitor self-renewal and differentiation throughout various stages of development. Furthermore, it is becoming apparent that TEAD proteins interact with other co-factors that influence stem cell biology. This Primer provides an overview of the role of TEAD proteins during development, focusing on their role in Hippo signalling as well as within other developmental, homeostatic and disease contexts.


Asunto(s)
Susceptibilidad a Enfermedades , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Familia de Multigenes , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Homeostasis , Humanos , Terapia Molecular Dirigida , Especificidad de Órganos , Regeneración , Especificidad de la Especie , Células Madre/citología , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Vertebrados
5.
Bioinformatics ; 39(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37449901

RESUMEN

MOTIVATION: Identification of cell types using single-cell RNA-seq is revolutionizing the study of multicellular organisms. However, typical single-cell RNA-seq analysis often involves post hoc manual curation to ensure clusters are transcriptionally distinct, which is time-consuming, error-prone, and irreproducible. RESULTS: To overcome these obstacles, we developed Cytocipher, a bioinformatics method and scverse compatible software package that statistically determines significant clusters. Application of Cytocipher to normal tissue, development, disease, and large-scale atlas data reveals the broad applicability and power of Cytocipher to generate biological insights in numerous contexts. This included the identification of cell types not previously described in the datasets analysed, such as CD8+ T cell subtypes in human peripheral blood mononuclear cells; cell lineage intermediate states during mouse pancreas development; and subpopulations of luminal epithelial cells over-represented in prostate cancer. Cytocipher also scales to large datasets with high-test performance, as shown by application to the Tabula Sapiens Atlas representing >480 000 cells. Cytocipher is a novel and generalizable method that statistically determines transcriptionally distinct and programmatically reproducible clusters from single-cell data. AVAILABILITY AND IMPLEMENTATION: The software version used for this manuscript has been deposited on Zenodo (https://doi.org/10.5281/zenodo.8089546), and is also available via github (https://github.com/BradBalderson/Cytocipher).


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica , Animales , Ratones , Humanos , Análisis de Secuencia de ARN/métodos , Perfilación de la Expresión Génica/métodos , Leucocitos Mononucleares , Análisis de Expresión Génica de una Sola Célula , Análisis de la Célula Individual , Programas Informáticos
6.
PLoS Genet ; 17(10): e1009334, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34710087

RESUMEN

Homozygous nonsense mutations in CEP55 are associated with several congenital malformations that lead to perinatal lethality suggesting that it plays a critical role in regulation of embryonic development. CEP55 has previously been studied as a crucial regulator of cytokinesis, predominantly in transformed cells, and its dysregulation is linked to carcinogenesis. However, its molecular functions during embryonic development in mammals require further investigation. We have generated a Cep55 knockout (Cep55-/-) mouse model which demonstrated preweaning lethality associated with a wide range of neural defects. Focusing our analysis on the neocortex, we show that Cep55-/- embryos exhibited depleted neural stem/progenitor cells in the ventricular zone as a result of significantly increased cellular apoptosis. Mechanistically, we demonstrated that Cep55-loss downregulates the pGsk3ß/ß-Catenin/Myc axis in an Akt-dependent manner. The elevated apoptosis of neural stem/progenitors was recapitulated using Cep55-deficient human cerebral organoids and we could rescue the phenotype by inhibiting active Gsk3ß. Additionally, we show that Cep55-loss leads to a significant reduction of ciliated cells, highlighting a novel role in regulating ciliogenesis. Collectively, our findings demonstrate a critical role of Cep55 during brain development and provide mechanistic insights that may have important implications for genetic syndromes associated with Cep55-loss.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neocórtex/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Carcinogénesis/metabolismo , Células Cultivadas , Citocinesis/fisiología , Homocigoto , Humanos , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Fenotipo
7.
Semin Cell Dev Biol ; 112: 61-68, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32771376

RESUMEN

Within the adult mammalian central nervous system, the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles houses neural stem cells (NSCs) that continue to produce neurons throughout life. Developmentally, the V-SVZ neurogenic niche arises during corticogenesis following the terminal differentiation of telencephalic radial glial cells (RGCs) into either adult neural stem cells (aNSCs) or ependymal cells. In mice, these two cellular populations form rosettes during the late embryonic and early postnatal period, with ependymal cells surrounding aNSCs. These aNSCs and ependymal cells serve a number of key purposes, including the generation of neurons throughout life (aNSCs), and acting as a barrier between the CSF and the parenchyma and promoting CSF bulk flow (ependymal cells). Interestingly, the development of this neurogenic niche, as well as its ongoing function, has been shown to be reliant on different aspects of lipid biology. In this review we discuss the developmental origins of the rodent V-SVZ neurogenic niche, and highlight research which has implicated a role for lipids in the physiology of this part of the brain. We also discuss the role of lipids in the maintenance of the V-SVZ niche, and discuss new research which has suggested that alterations to lipid biology could contribute to ependymal cell dysfunction in aging and disease.


Asunto(s)
Envejecimiento/genética , Epéndimo/metabolismo , Lípidos/genética , Células-Madre Neurales/metabolismo , Envejecimiento/patología , Animales , Proliferación Celular/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Epéndimo/crecimiento & desarrollo , Epéndimo/patología , Humanos , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Ratones , Células-Madre Neurales/fisiología , Neurogénesis/genética , Neuronas/metabolismo , Neuronas/patología , Telencéfalo/metabolismo , Telencéfalo/patología
8.
Hum Mol Genet ; 29(2): 248-263, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31816041

RESUMEN

WDR62 mutations that result in protein loss, truncation or single amino-acid substitutions are causative for human microcephaly, indicating critical roles in cell expansion required for brain development. WDR62 missense mutations that retain protein expression represent partial loss-of-function mutants that may therefore provide specific insights into radial glial cell processes critical for brain growth. Here we utilized CRISPR/Cas9 approaches to generate three strains of WDR62 mutant mice; WDR62 V66M/V66M and WDR62R439H/R439H mice recapitulate conserved missense mutations found in humans with microcephaly, with the third strain being a null allele (WDR62stop/stop). Each of these mutations resulted in embryonic lethality to varying degrees and gross morphological defects consistent with ciliopathies (dwarfism, anophthalmia and microcephaly). We find that WDR62 mutant proteins (V66M and R439H) localize to the basal body but fail to recruit CPAP. As a consequence, we observe deficient recruitment of IFT88, a protein that is required for cilia formation. This underpins the maintenance of radial glia as WDR62 mutations caused premature differentiation of radial glia resulting in reduced generation of neurons and cortical thinning. These findings highlight the important role of the primary cilium in neocortical expansion and implicate ciliary dysfunction as underlying the pathology of MCPH2 patients.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cilios/metabolismo , Ciliopatías/genética , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Anoftalmos/embriología , Anoftalmos/genética , Anoftalmos/metabolismo , Apoptosis/genética , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cilios/genética , Cilios/patología , Ciliopatías/embriología , Ciliopatías/metabolismo , Ciliopatías/patología , Enanismo/embriología , Enanismo/genética , Enanismo/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Fibroblastos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcefalia/embriología , Microcefalia/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Mutación Missense , Neocórtex/embriología , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas Supresoras de Tumor/genética
9.
Development ; 146(18)2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31488566

RESUMEN

During development, the p75 neurotrophin receptor (p75NTR) is widely expressed in the nervous system where it regulates neuronal differentiation, migration and axonal outgrowth. p75NTR also mediates the survival and death of newly born neurons, with functional outcomes being dependent on both timing and cellular context. Here, we show that knockout of p75NTR from embryonic day 10 (E10) in neural progenitors using a conditional Nestin-Cre p75NTR floxed mouse causes increased apoptosis of progenitor cells. By E14.5, the number of Tbr2-positive progenitor cells was significantly reduced and the rate of neurogenesis was halved. Furthermore, in adult knockout mice, there were fewer cortical pyramidal neurons, interneurons, cholinergic basal forebrain neurons and striatal neurons, corresponding to a relative reduction in volume of these structures. Thalamic midline fusion during early postnatal development was also impaired in Nestin-Cre p75NTR floxed mice, indicating a novel role for p75NTR in the formation of this structure. The phenotype of this strain demonstrates that p75NTR regulates multiple aspects of brain development, including cortical progenitor cell survival, and that expression during early neurogenesis is required for appropriate formation of telencephalic structures.


Asunto(s)
Prosencéfalo Basal/embriología , Neocórtex/embriología , Neostriado/embriología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Tálamo/embriología , Animales , Animales Recién Nacidos , Caspasa 3/metabolismo , Proliferación Celular , Supervivencia Celular , Aparato de Golgi/metabolismo , Interneuronas/metabolismo , Ratones , Nestina/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Tamaño de los Órganos , Células Piramidales/metabolismo
10.
Biol Reprod ; 106(6): 1191-1205, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35243487

RESUMEN

Members of the nuclear factor I (NFI) family are key regulators of stem cell biology during development, with well-documented roles for NFIA, NFIB, and NFIX in a variety of developing tissues, including brain, muscle, and lung. Given the central role these factors play in stem cell biology, we posited that they may be pivotal for spermatogonial stem cells or further developing spermatogonia during testicular development. Surprisingly, in stark contrast to other developing organ systems where NFI members are co-expressed, these NFI family members show discrete patterns of expression within the seminiferous tubules. Sertoli cells (spermatogenic supporting cells) express NFIA, spermatocytes express NFIX, round spermatids express NFIB, and peritubular myoid cells express each of these three family members. Further analysis of NFIX expression during the cycle of the seminiferous epithelium revealed expression not in spermatogonia, as we anticipated, but in spermatocytes. These data suggested a potential role for NFIX in spermatogenesis. To investigate, we analyzed mice with constitutive deletion of Nfix (Nfix-null). Assessment of germ cells in the postnatal day 20 (P20) testes of Nfix-null mice revealed that spermatocytes initiate meiosis, but zygotene stage spermatocytes display structural defects in the synaptonemal complex, and increased instances of unrepaired DNA double-strand breaks. Many developing spermatocytes in the Nfix-null testis exhibited multinucleation. As a result of these defects, spermatogenesis is blocked at early diplotene and very few round spermatids are produced. Collectively, these novel data establish the global requirement for NFIX in correct meiotic progression during the first wave of spermatogenesis.


Asunto(s)
Factores de Transcripción NFI , Espermatogonias , Testículo , Animales , Masculino , Meiosis , Ratones , Ratones Noqueados , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Espermatocitos/metabolismo , Espermatogénesis/genética , Testículo/metabolismo
11.
Mol Psychiatry ; 26(11): 6880-6895, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34059796

RESUMEN

Neural stem cells in the human subependymal zone (SEZ) generate neuronal progenitor cells that can differentiate and integrate as inhibitory interneurons into cortical and subcortical brain regions; yet the extent of adult neurogenesis remains unexplored in schizophrenia and bipolar disorder. We verified the existence of neurogenesis across the lifespan by chartering transcriptional alterations (2 days-103 years, n = 70) and identifying cells indicative of different stages of neurogenesis in the human SEZ. Expression of most neural stem and neuronal progenitor cell markers decreased during the first postnatal years and remained stable from childhood into ageing. We next discovered reduced neural stem and neuronal progenitor cell marker expression in the adult SEZ in schizophrenia and bipolar disorder compared to controls (n = 29-32 per group). RNA sequencing identified increased expression of the macrophage marker CD163 as the most significant molecular change in schizophrenia. CD163+ macrophages, which were localised along blood vessels and in the parenchyma within 10 µm of neural stem and progenitor cells, had increased density in schizophrenia but not in bipolar disorder. Macrophage marker expression negatively correlated with neuronal progenitor marker expression in schizophrenia but not in controls or bipolar disorder. Reduced neurogenesis and increased macrophage marker expression were also associated with polygenic risk for schizophrenia. Our results support that the human SEZ retains the capacity to generate neuronal progenitor cells throughout life, although this capacity is limited in schizophrenia and bipolar disorder. The increase in macrophages in schizophrenia but not in bipolar disorder indicates that immune cells may impair neurogenesis in the adult SEZ in a disease-specific manner.


Asunto(s)
Células-Madre Neurales , Esquizofrenia , Adulto , Niño , Humanos , Macrófagos , Neurogénesis/fisiología , Neuronas
12.
Cereb Cortex ; 31(3): 1763-1775, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33188399

RESUMEN

Genetic association studies have identified many factors associated with neurodevelopmental disorders such as autism spectrum disorder (ASD). However, the way these genes shape neuroanatomical structure and connectivity is poorly understood. Recent research has focused on proteins that act as points of convergence for multiple factors, as these may provide greater insight into understanding the biology of neurodevelopmental disorders. USP9X, a deubiquitylating enzyme that regulates the stability of many ASD-related proteins, is one such point of convergence. Loss of function variants in human USP9X lead to brain malformations, which manifest as a neurodevelopmental syndrome that frequently includes ASD, but the underlying structural and connectomic abnormalities giving rise to patient symptoms is unknown. Here, we analyzed forebrain-specific Usp9x knockout mice (Usp9x-/y) to address this knowledge gap. Usp9x-/y mice displayed abnormal communication and social interaction behaviors. Moreover, the absence of Usp9x culminated in reductions to the size of multiple brain regions. Diffusion tensor magnetic resonance imaging revealed deficits in all three major forebrain commissures, as well as long-range hypoconnectivity between cortical and subcortical regions. These data identify USP9X as a key regulator of brain formation and function, and provide insights into the neurodevelopmental syndrome arising as a consequence of USP9X mutations in patients.


Asunto(s)
Corteza Cerebral/fisiopatología , Vías Nerviosas/fisiopatología , Neurogénesis/fisiología , Ubiquitina Tiolesterasa/metabolismo , Animales , Conducta Animal , Masculino , Ratones , Ratones Noqueados
13.
Bioessays ; 42(10): e2000065, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32767425

RESUMEN

What is the function of new neurons entering the olfactory bulb? Many insights regarding the molecular control of adult neurogenesis have been uncovered, but the purpose of new neurons entering the olfactory bulb has been difficult to ascertain. Here, studies investigating the role of adult neurogenesis in olfactory discrimination in mice are reviewed. Studies in which adult neurogenesis is affected are highlighted, with a focus on the role of environment enrichment and what happens during ageing. There is evidence for a role of adult neurogenesis in fine discrimination tasks, as underscored by studies that enhance adult neurogenesis. This is also observed in ageing studies, where older mice with reduced levels of adult neurogenesis perform poorly in olfactory discrimination. Differences in methodology that could account for alternative conclusions, and the importance of specificity in methods being used to investigate the effect of adult neurogenesis in olfactory performance are emphasized.


Asunto(s)
Neurogénesis , Bulbo Olfatorio , Animales , Ratones , Neuronas , Olfato
14.
Genomics ; 113(4): 1855-1866, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33878366

RESUMEN

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) is the primary protocol for detecting genome-wide DNA-protein interactions, and therefore a key tool for understanding transcriptional regulation. A number of factors, including low specificity of antibody and cellular heterogeneity of sample, may cause "peak" callers to output noise and experimental artefacts. Statistically combining multiple experimental replicates from the same condition could significantly enhance our ability to distinguish actual transcription factor binding events, even when peak caller accuracy and consistency of detection are compromised. We adapted the rank-product test to statistically evaluate the reproducibility from any number of ChIP-seq experimental replicates. We demonstrate over a number of benchmarks that our adaptation "ChIP-R" (pronounced 'chipper') performs as well as or better than comparable approaches on recovering transcription factor binding sites in ChIP-seq peak data. We also show ChIP-R extends to evaluate ATAC-seq peaks, finding reproducible peak sets even at low sequencing depth. ChIP-R decomposes peaks across replicates into "fragments" which either form part of a peak in a replicate, or not. We show that by re-analysing existing data sets, ChIP-R reconstructs reproducible peaks from fragments with enhanced biological enrichment relative to current strategies.


Asunto(s)
Algoritmos , Secuenciación de Inmunoprecipitación de Cromatina , Sitios de Unión , Inmunoprecipitación de Cromatina/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/métodos
15.
Development ; 145(3)2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437824

RESUMEN

Our understanding of the transcriptional programme underpinning adult hippocampal neurogenesis is incomplete. In mice, under basal conditions, adult hippocampal neural stem cells (AH-NSCs) generate neurons and astrocytes, but not oligodendrocytes. The factors limiting oligodendrocyte production, however, remain unclear. Here, we reveal that the transcription factor NFIX plays a key role in this process. NFIX is expressed by AH-NSCs, and its expression is sharply upregulated in adult hippocampal neuroblasts. Conditional ablation of Nfix from AH-NSCs, coupled with lineage tracing, transcriptomic sequencing and behavioural studies collectively reveal that NFIX is cell-autonomously required for neuroblast maturation and survival. Moreover, a small number of AH-NSCs also develop into oligodendrocytes following Nfix deletion. Remarkably, when Nfix is deleted specifically from intermediate progenitor cells and neuroblasts using a Dcx-creERT2 driver, these cells also display elevated signatures of oligodendrocyte gene expression. Together, these results demonstrate the central role played by NFIX in neuroblasts within the adult hippocampal stem cell neurogenic niche in promoting the maturation and survival of these cells, while concomitantly repressing oligodendrocyte gene expression signatures.


Asunto(s)
Hipocampo/citología , Hipocampo/metabolismo , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular , Proteína Doblecortina , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Nicho de Células Madre/genética , Nicho de Células Madre/fisiología , Regulación hacia Arriba
16.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23964093

RESUMEN

The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC quiescence have been identified, the transcriptional mechanisms involved are mostly unknown, largely due to lack of a cell culture model. In this study, we first demonstrate that NSC cultures (NS cells) exposed to BMP4 acquire cellular and transcriptional characteristics of quiescent cells. We then use epigenomic profiling to identify enhancers associated with the quiescent NS cell state. Motif enrichment analysis of these enhancers predicts a major role for the nuclear factor one (NFI) family in the gene regulatory network controlling NS cell quiescence. Interestingly, we found that the family member NFIX is robustly induced when NS cells enter quiescence. Using genome-wide location analysis and overexpression and silencing experiments, we demonstrate that NFIX has a major role in the induction of quiescence in cultured NSCs. Transcript profiling of NS cells overexpressing or silenced for Nfix and the phenotypic analysis of the hippocampus of Nfix mutant mice suggest that NFIX controls the quiescent state by regulating the interactions of NSCs with their microenvironment.


Asunto(s)
Epigénesis Genética , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Proteína Morfogenética Ósea 4/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Factores de Transcripción NFI/genética , Células-Madre Neurales/efectos de los fármacos , Unión Proteica
17.
Cerebellum ; 19(1): 89-101, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31838646

RESUMEN

Transcriptional regulation plays a central role in controlling neural stem and progenitor cell proliferation and differentiation during neurogenesis. For instance, transcription factors from the nuclear factor I (NFI) family have been shown to co-ordinate neural stem and progenitor cell differentiation within multiple regions of the embryonic nervous system, including the neocortex, hippocampus, spinal cord and cerebellum. Knockout of individual Nfi genes culminates in similar phenotypes, suggestive of common target genes for these transcription factors. However, whether or not the NFI family regulates common suites of genes remains poorly defined. Here, we use granule neuron precursors (GNPs) of the postnatal murine cerebellum as a model system to analyse regulatory targets of three members of the NFI family: NFIA, NFIB and NFIX. By integrating transcriptomic profiling (RNA-seq) of Nfia- and Nfix-deficient GNPs with epigenomic profiling (ChIP-seq against NFIA, NFIB and NFIX, and DNase I hypersensitivity assays), we reveal that these transcription factors share a large set of potential transcriptional targets, suggestive of complementary roles for these NFI family members in promoting neural development.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Animales Recién Nacidos , Cerebelo/citología , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/genética , Neurogénesis/fisiología , Embarazo
18.
Gastrointest Endosc ; 91(4): 882-893.e4, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31715173

RESUMEN

BACKGROUND AND AIMS: Gastroenterology fellowships need to ensure that trainees achieve competence in upper endoscopy (EGD) and colonoscopy. Because the impact of structured feedback remains unknown in endoscopy training, this study compared the effect of structured feedback with standard feedback on trainee learning curves for EGD and colonoscopy. METHODS: In this multicenter, cluster, randomized controlled trial, trainees received either individualized quarterly learning curves or feedback standard to their fellowship. Assessment was performed in all trainees using the Assessment of Competency in Endoscopy tool on 5 consecutive procedures after every 25 EGDs and colonoscopies. Individual learning curves were created using cumulative sum (CUSUM) analysis. The primary outcome was the mean CUSUM score in overall technical and overall cognitive skills. RESULTS: In all, 13 programs including 132 trainees participated. The intervention arm (6 programs, 51 trainees) contributed 558 EGD and 600 colonoscopy assessments. The control arm (7 programs, 81 trainees) provided 305 EGD and 468 colonoscopy assessments. For EGD, the intervention arm (-.7 [standard deviation {SD}, 1.3]) had a superior mean CUSUM score in overall cognitive skills compared with the control arm (1.6 [SD, .8], P = .03) but not in overall technical skills (intervention, -.26 [SD, 1.4]; control, 1.76 [SD, .7]; P = .06). For colonoscopy, no differences were found between the 2 arms in overall cognitive skills (intervention, -.7 [SD, 1.3]; control, .7 [SD, 1.3]; P = .95) or overall technical skills (intervention, .1 [SD, 1.5]; control, -.1 [SD, 1.5]; P = .77). CONCLUSIONS: Quarterly feedback in the form of individualized learning curves did not affect learning curves for EGD and colonoscopy in a clinically meaningful manner. (Clinical trial registration number: NCT02891304.).


Asunto(s)
Curva de Aprendizaje , Competencia Clínica , Colonoscopía , Retroalimentación , Gastroenterología/educación , Humanos
19.
Cereb Cortex ; 29(8): 3590-3604, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-30272140

RESUMEN

Understanding the migration of newborn neurons within the brain presents a major challenge in contemporary biology. Neuronal migration is widespread within the developing brain but is also important within the adult brain. For instance, stem cells within the ventricular-subventricular zone (V-SVZ) and the subgranular zone of dentate gyrus of the adult rodent brain produce neuroblasts that migrate to the olfactory bulb and granule cell layer of the dentate gyrus, respectively, where they regulate key brain functions including innate olfactory responses, learning, and memory. Critically, our understanding of the factors mediating neuroblast migration remains limited. The transcription factor nuclear factor I X (NFIX) has previously been implicated in embryonic cortical development. Here, we employed conditional ablation of Nfix from the adult mouse brain and demonstrated that the removal of this gene from either neural stem and progenitor cells, or neuroblasts, within the V-SVZ culminated in neuroblast migration defects. Mechanistically, we identified aberrant neuroblast branching, due in part to increased expression of the guanylyl cyclase natriuretic peptide receptor 2 (Npr2), as a factor contributing to abnormal migration in Nfix-deficient adult mice. Collectively, these data provide new insights into how neuroblast migration is regulated at a transcriptional level within the adult brain.


Asunto(s)
Movimiento Celular/genética , Giro Dentado/citología , Ventrículos Laterales/citología , Factores de Transcripción NFI/genética , Células-Madre Neurales/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ratones , Células-Madre Neurales/citología , Neurogénesis/genética , Receptores del Factor Natriurético Atrial/genética
20.
Am J Med Genet C Semin Med Genet ; 181(4): 611-626, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31730271

RESUMEN

The nuclear factor one (NFI) site-specific DNA-binding proteins represent a family of transcription factors that are important for the development of multiple organ systems, including the brain. During brain development in mice, the expression patterns of Nfia, Nfib, and Nfix overlap, and knockout mice for each of these exhibit overlapping brain defects, including megalencephaly, dysgenesis of the corpus callosum, and enlarged ventricles, which implies a common but not redundant function in brain development. In line with these models, human phenotypes caused by haploinsufficiency of NFIA, NFIB, and NFIX display significant overlap, sharing neurodevelopmental deficits, macrocephaly, brain anomalies, and variable somatic overgrowth. Other anomalies may be present depending on the NFI gene involved. The possibility of variants in NFI genes should therefore be considered in individuals with intellectual disability and brain overgrowth, with individual NFI-related conditions being differentiated from one another by additional signs and symptoms. The exception is provided by specific NFIX variants that act in a dominant negative manner, as these cause a recognizable entity with more severe cognitive impairment and marked bone dysplasia, Marshall-Smith syndrome. NFIX duplications are associated with a phenotype opposite to that of haploinsufficiency, characterized by short stature, small head circumference, and delayed bone age. The spectrum of NFI-related disorders will likely be further expanded, as larger cohorts are assessed.


Asunto(s)
Crecimiento/genética , Mutación , Factores de Transcripción NFI/genética , Anomalías Múltiples/genética , Animales , Enfermedades del Desarrollo Óseo/genética , Anomalías Craneofaciales/genética , Duplicación de Gen , Trastornos del Crecimiento/genética , Humanos , Ratones , Displasia Septo-Óptica/genética , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA