Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biomaterials ; 283: 121416, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35217483

RESUMEN

Personalised nanomedicine is an advancing field which has developed significant improvements for targeting therapeutics to aggressive cancer and with fewer side effects. The treatment of gliomas such as glioblastoma (or other brain tumours), with nanomedicine is complicated by a commonly poor accumulation of drugs in tumour tissue owing to the partially intact blood-brain barrier (BBB). Nonetheless, the BBB becomes compromised following surgical intervention, and gradually with disease progression. Increased vasculature permeability generated by a tumour, combined with decreased BBB integrity, offers a mechanism to enhance therapeutic outcomes. We monitored a spontaneous glioma tumour model in immunocompetent mice with ongoing T2-weighted and contrast-enhanced T1-weighted magnetic resonance imaging gradient echo and spin echo sequences to predict an optimal "leakiness" stage for nanomedicine injections. To ascertain the effectiveness of targeted nanomedicines in treating brain tumours, subsequent systemic administration of targeted hyperbranched polymers was then utislised, to deliver the therapeutic payload when both the tumour and brain vascularity had become sufficiently susceptible to allow drug accumulation. Treatment with either doxorubicin-loaded hyperbranched polymer, or the same nanomedicine targeted to an ephrin receptor (EphA2) using a bispecific antibody, resulted in uptake of chemotherapeutic doxorubicin in the tumour and in reduced tumour growth. Compared to vehicle and doxorubicin only, nanoparticle delivered doxorubicin resulted in increased tumour apoptosis, while averting cardiotoxicity. This suggests that polyethylene based (PEGylated)-nanoparticle delivered doxorubicin could provide a more efficient treatment in tumours with a disrupted BBB, and that treatment should commence immediately following detection of gadolinium permeability, with early detection and ongoing 'leakiness' monitoring in susceptible patients being a key factor.


Asunto(s)
Neoplasias Encefálicas , Nanomedicina , Animales , Barrera Hematoencefálica , Encéfalo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina , Sistemas de Liberación de Medicamentos/métodos , Humanos , Ratones , Nanomedicina/métodos
2.
Acta Neuropathol ; 143(4): 427-451, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35169893

RESUMEN

Pediatric neoplasms in the central nervous system (CNS) are the leading cause of cancer-related deaths in children. Recent developments in molecular analyses have greatly contributed to a more accurate diagnosis and risk stratification of CNS tumors. Additionally, sequencing studies have identified various, often entity specific, tumor-driving events. In contrast to adult tumors, which often harbor multiple mutated oncogenic drivers, the number of mutated genes in pediatric cancers is much lower and many tumors can have a single oncogenic driver. Moreover, in children, much more than in adults, fusion proteins play an important role in driving tumorigenesis, and many different fusions have been identified as potential driver events in pediatric CNS neoplasms. However, a comprehensive overview of all the different reported oncogenic fusion proteins in pediatric CNS neoplasms is still lacking. A better understanding of the fusion proteins detected in these tumors and of the molecular mechanisms how these proteins drive tumorigenesis, could improve diagnosis and further benefit translational research into targeted therapies necessary to treat these distinct entities. In this review, we discuss the different oncogenic fusions reported in pediatric CNS neoplasms and their structure to create an overview of the variety of oncogenic fusion proteins to date, the tumor entities they occur in and their proposed mode of action.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Fusión de Oncogenes , Adulto , Carcinogénesis , Neoplasias del Sistema Nervioso Central/genética , Niño , Humanos , Fusión de Oncogenes/genética , Proteínas de Fusión Oncogénica/genética
3.
Elife ; 102021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33945466

RESUMEN

Corpus callosum dysgenesis (CCD) is a congenital disorder that incorporates either partial or complete absence of the largest cerebral commissure. Remodelling of the interhemispheric fissure (IHF) provides a substrate for callosal axons to cross between hemispheres, and its failure is the main cause of complete CCD. However, it is unclear whether defects in this process could give rise to the heterogeneity of expressivity and phenotypes seen in human cases of CCD. We identify incomplete IHF remodelling as the key structural correlate for the range of callosal abnormalities in inbred and outcrossed BTBR mouse strains, as well as in humans with partial CCD. We identify an eight base-pair deletion in Draxin and misregulated astroglial and leptomeningeal proliferation as genetic and cellular factors for variable IHF remodelling and CCD in BTBR strains. These findings support a model where genetic events determine corpus callosum structure by influencing leptomeningeal-astroglial interactions at the IHF.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Cuerpo Calloso/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Adulto , Anciano , Agenesia del Cuerpo Calloso/patología , Animales , Estudios de Cohortes , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/patología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fenotipo , Adulto Joven
4.
Carcinogenesis ; 42(3): 357-368, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33346791

RESUMEN

Nuclear factor one (NFI) transcription factors are implicated in both brain development and cancer in mice and humans and play an essential role in glial differentiation. NFI expression is reduced in human astrocytoma samples, particularly those of higher grade, whereas over-expression of NFI protein can induce the differentiation of glioblastoma cells within human tumour xenografts and in glioblastoma cell lines in vitro. These data indicate that NFI proteins may act as tumour suppressors in glioma. To test this hypothesis, we generated complex mouse genetic crosses involving six alleles to target gene deletion of known tumour suppressor genes that induce endogenous high-grade glioma in mice, and overlaid this with loss of function Nfi mutant alleles, Nfia and Nfib, a reporter transgene and an inducible Cre allele. Deletion of Nfi resulted in reduced survival time of the mice, increased tumour load and a more aggressive tumour phenotype than observed in glioma mice with normal expression of NFI. Together, these data indicate that NFI genes represent a credible target for both diagnostic analyses and therapeutic strategies to combat high-grade glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Factores de Transcripción NFI/metabolismo , Animales , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción NFI/genética
5.
ACS Cent Sci ; 6(5): 727-738, 2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32490189

RESUMEN

Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.

6.
Neuroimage ; 217: 116868, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32360691

RESUMEN

Corpus callosum dysgenesis (CCD) describes a collection of brain malformations in which the main fiber tract connecting the two hemispheres is either absent (complete CCD, or 'agenesis of the corpus callosum') or reduced in size (partial CCD). Humans with these neurodevelopmental disorders have a wide range of cognitive outcomes, including seemingly preserved features of interhemispheric communication in some cases. However, the structural substrates that could underlie this variability in outcome remain to be fully elucidated. Here, for the first time, we characterize the global brain connectivity of a mouse model of complete and partial CCD. We demonstrate features of structural brain connectivity that model those predicted in humans with CCD, including Probst bundles in complete CCD and heterotopic sigmoidal connections in partial CCD. Crucially, we also histologically validate the recently predicted ectopic sigmoid bundle present in humans with partial CCD, validating the utility of this mouse model for fine anatomical studies of this disorder. Taken together, this work describes a mouse model of altered structural connectivity in variable severity CCD and forms a foundation for future studies investigating the function and mechanisms of development of plastic tracts in developmental disorders of brain connectivity.


Asunto(s)
Agenesia del Cuerpo Calloso/diagnóstico por imagen , Vías Nerviosas/diagnóstico por imagen , Animales , Conectoma , Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Electroporación , Femenino , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Plasticidad Neuronal , Embarazo
7.
Neuron ; 106(3): 404-420.e8, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32135084

RESUMEN

De novo germline mutations in the RNA helicase DDX3X account for 1%-3% of unexplained intellectual disability (ID) cases in females and are associated with autism, brain malformations, and epilepsy. Yet, the developmental and molecular mechanisms by which DDX3X mutations impair brain function are unknown. Here, we use human and mouse genetics and cell biological and biochemical approaches to elucidate mechanisms by which pathogenic DDX3X variants disrupt brain development. We report the largest clinical cohort to date with DDX3X mutations (n = 107), demonstrating a striking correlation between recurrent dominant missense mutations, polymicrogyria, and the most severe clinical outcomes. We show that Ddx3x controls cortical development by regulating neuron generation. Severe DDX3X missense mutations profoundly disrupt RNA helicase activity, induce ectopic RNA-protein granules in neural progenitors and neurons, and impair translation. Together, these results uncover key mechanisms underlying DDX3X syndrome and highlight aberrant RNA metabolism in the pathogenesis of neurodevelopmental disease.


Asunto(s)
Corteza Cerebral/metabolismo , ARN Helicasas DEAD-box/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Neurogénesis , Animales , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/anomalías , Corteza Cerebral/embriología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos del Neurodesarrollo/patología , ARN/metabolismo
8.
J Neurooncol ; 146(1): 41-53, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31760595

RESUMEN

INTRODUCTION: Malignant astrocytomas are composed of heterogeneous cell populations. Compared to grade IV glioblastoma, low-grade astrocytomas have more differentiated cells and are associated with a better prognosis. Therefore, inducing cellular differentiation to alter the behaviour of high-grade astrocytomas may serve as a therapeutic strategy. The nuclear factor one (NFI) transcription factors are essential for normal astrocytic differentiation. Here, we investigate whether family members NFIA and NFIB act as effectors of cellular differentiation in glioblastoma. METHODS: We analysed expression of NFIA and NFIB in mRNA expression data of high-grade astrocytoma and with immunofluorescence co-staining. Furthermore, we induced NFI expression in patient-derived subcutaneous glioblastoma xenografts via in vivo electroporation. RESULTS: The expression of NFIA and NFIB is reduced in glioblastoma as compared to lower grade astrocytomas. At a cellular level, their expression is associated with differentiated and mature astrocyte-like tumour cells. In vivo analyses consistently demonstrate that expression of either NFIA or NFIB is sufficient to promote tumour cell differentiation in glioblastoma xenografts. CONCLUSION: Our findings indicate that both NFIA and NFIB may have an endogenous pro-differentiative function in astrocytomas, similar to their role in normal astrocyte differentiation. Overall, our study establishes a basis for further investigation of targeting NFI-mediated differentiation as a potential differentiation therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Glioblastoma/patología , Factores de Transcripción NFI/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Transcripción NFI/genética , Clasificación del Tumor , Neurogénesis , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
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
10.
Nat Commun ; 10(1): 3914, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477715

RESUMEN

YAP1 fusion-positive supratentorial ependymomas predominantly occur in infants, but the molecular mechanisms of oncogenesis are unknown. Here we show YAP1-MAMLD1 fusions are sufficient to drive malignant transformation in mice, and the resulting tumors share histo-molecular characteristics of human ependymomas. Nuclear localization of YAP1-MAMLD1 protein is mediated by MAMLD1 and independent of YAP1-Ser127 phosphorylation. Chromatin immunoprecipitation-sequencing analyses of human YAP1-MAMLD1-positive ependymoma reveal enrichment of NFI and TEAD transcription factor binding site motifs in YAP1-bound regulatory elements, suggesting a role for these transcription factors in YAP1-MAMLD1-driven tumorigenesis. Mutation of the TEAD binding site in the YAP1 fusion or repression of NFI targets prevents tumor induction in mice. Together, these results demonstrate that the YAP1-MAMLD1 fusion functions as an oncogenic driver of ependymoma through recruitment of TEADs and NFIs, indicating a rationale for preclinical studies to block the interaction between YAP1 fusions and NFI and TEAD transcription factors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Encefálicas/metabolismo , Carcinogénesis/metabolismo , Proteínas de Unión al ADN/metabolismo , Ependimoma/metabolismo , Factores de Transcripción NFI/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/genética , Ependimoma/genética , Ependimoma/patología , Células HEK293 , Humanos , Ratones , Factores de Transcripción NFI/genética , Células 3T3 NIH , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
11.
Am J Hum Genet ; 103(5): 752-768, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388402

RESUMEN

The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.


Asunto(s)
Haploinsuficiencia/genética , Discapacidad Intelectual/genética , Megalencefalia/genética , Factores de Transcripción NFI/genética , Adolescente , Adulto , Animales , Corteza Cerebral/patología , Niño , Preescolar , Codón sin Sentido/genética , Estudios de Cohortes , Cuerpo Calloso/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple/genética , Adulto Joven
12.
Dev Biol ; 432(2): 286-297, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29106906

RESUMEN

During mouse spinal cord development, ventricular zone progenitor cells transition from producing neurons to producing glia at approximately embryonic day 11.5, a process known as the gliogenic switch. The transcription factors Nuclear Factor I (NFI) A and B initiate this developmental transition, but the contribution of a third NFI member, NFIX, remains unknown. Here, we reveal that ventricular zone progenitor cells within the spinal cord express NFIX after the onset of NFIA and NFIB expression, and after the gliogenic switch has occurred. Mice lacking NFIX exhibit normal neurogenesis within the spinal cord, and, while early astrocytic differentiation proceeds normally, aspects of terminal astrocytic differentiation are impaired. Finally, we report that, in the absence of Nfia or Nfib, there is a marked reduction in the spinal cord expression of NFIX, and that NFIB can transcriptionally activate Nfix expression in vitro. These data demonstrate that NFIX is part of the downstream transcriptional program through which NFIA and NFIB coordinate gliogenesis within the spinal cord. This hierarchical organisation of NFI protein expression and function during spinal cord gliogenesis reveals a previously unrecognised auto-regulatory mechanism within this gene family.


Asunto(s)
Factores de Transcripción NFI/metabolismo , Médula Espinal/embriología , Animales , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFI/genética , Neurogénesis , Neuroglía/metabolismo , Neuronas/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Células Madre/metabolismo , Activación Transcripcional
13.
Cancer Lett ; 410: 124-138, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28962832

RESUMEN

The nuclear factor I (NFI) transcription factors play important roles during normal development and have been associated with developmental abnormalities in humans. All four family members, NFIA, NFIB, NFIC and NFIX, have a homologous DNA binding domain and function by regulating cell proliferation and differentiation via the transcriptional control of their target genes. More recently, NFI genes have also been implicated in cancer based on genomic analyses and studies of animal models in a variety of tumours across multiple organ systems. However, the association between their functions in development and in cancer is not well described. In this review, we summarise the evidence suggesting a converging role for the NFI genes in development and cancer. Our review includes all cancer types in which the NFI genes are implicated, focusing predominantly on studies demonstrating their oncogenic or tumour-suppressive potential. We conclude by presenting the challenges impeding our understanding of NFI function in cancer biology, and demonstrate how a developmental perspective may contribute towards overcoming such hurdles.


Asunto(s)
Factores de Transcripción NFI/metabolismo , Neoplasias/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción NFI/genética , Neoplasias/genética , Neoplasias/patología , Transducción de Señal , Transcripción Genética
14.
J Comp Neurol ; 525(11): 2465-2483, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28295292

RESUMEN

The nuclear factor I (NFI) family of transcription factors plays an important role in the development of the cerebral cortex in humans and mice. Disruption of nuclear factor IA (NFIA), nuclear factor IB (NFIB), or nuclear factor IX (NFIX) results in abnormal development of the corpus callosum, lateral ventricles, and hippocampus. However, the expression or function of these genes has not been examined in detail in the adult brain, and the cell type-specific expression of NFIA, NFIB, and NFIX is currently unknown. Here, we demonstrate that the expression of each NFI protein shows a distinct laminar pattern in the adult mouse neocortex and that their cell type-specific expression differs depending on the family member. NFIA expression was more frequently observed in astrocytes and oligodendroglia, whereas NFIB expression was predominantly localized to astrocytes and neurons. NFIX expression was most commonly observed in neurons. The NFI proteins were equally distributed within microglia, and the ependymal cells lining the ventricles of the brain expressed all three proteins. In the hippocampus, the NFI proteins were expressed during all stages of neural stem cell differentiation in the dentate gyrus, with higher expression intensity in neuroblast cells as compared to quiescent stem cells and mature granule neurons. These findings suggest that the NFI proteins may play distinct roles in cell lineage specification or maintenance, and establish the basis for further investigation of their function in the adult brain and their emerging role in disease.


Asunto(s)
Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción NFI/biosíntesis , Neuroglía/metabolismo , Neuronas/metabolismo , Factores de Edad , Animales , Diferenciación Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Factores de Transcripción NFI/genética
15.
Nat Genet ; 49(4): 511-514, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28250454

RESUMEN

Brain malformations involving the corpus callosum are common in children with developmental disabilities. We identified DCC mutations in four families and five sporadic individuals with isolated agenesis of the corpus callosum (ACC) without intellectual disability. DCC mutations result in variable dominant phenotypes with decreased penetrance, including mirror movements and ACC associated with a favorable developmental prognosis. Possible phenotypic modifiers include the type and location of mutation and the sex of the individual.


Asunto(s)
Agenesia del Cuerpo Calloso/genética , Discapacidades del Desarrollo/genética , Mutación/genética , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/genética , Anomalías Múltiples/genética , Encéfalo/patología , Cuerpo Calloso/patología , Receptor DCC , Familia , Femenino , Humanos , Masculino , Malformaciones del Sistema Nervioso/genética , Células-Madre Neurales/patología , Penetrancia , Fenotipo
16.
Brain Neurosci Adv ; 1: 2398212817739433, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-32166136

RESUMEN

BACKGROUND: Nuclear factor I family members nuclear factor I A and nuclear factor I B play important roles during cerebral cortical development. Nuclear factor I A and nuclear factor I B regulate similar biological processes, as their expression patterns, regulation of target genes and individual knockout phenotypes overlap. We hypothesised that the combined allelic loss of Nfia and Nfib would culminate in more severe defects in the cerebral cortex than loss of a single member. METHODS: We combined immunofluorescence, co-immunoprecipitation, gene expression analysis and immunohistochemistry on knockout mouse models to investigate whether nuclear factor I A and nuclear factor I B function similarly and whether increasing allelic loss of Nfia and Nfib caused a more severe phenotype. RESULTS: We determined that the biological functions of nuclear factor I A and nuclear factor I B overlap during early cortical development. These proteins are co-expressed and can form heterodimers in vivo. Differentially regulated genes that are shared between Nfia and Nfib knockout mice are highly enriched for nuclear factor I binding sites in their promoters and are associated with neurodevelopment. We found that compound heterozygous deletion of both genes resulted in a cortical phenotype similar to that of single homozygous Nfia or Nfib knockout embryos. This was characterised by retention of the interhemispheric fissure, dysgenesis of the corpus callosum and a malformed dentate gyrus. Double homozygous knockout of Nfia and Nfib resulted in a more severe phenotype, with increased ventricular enlargement and decreased numbers of differentiated glia and neurons. CONCLUSION: In the developing cerebral cortex, nuclear factor I A and nuclear factor I B share similar biological functions and function additively, as the combined allelic loss of these genes directly correlates with the severity of the developmental brain phenotype.

17.
Cell Rep ; 17(3): 735-747, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27732850

RESUMEN

The corpus callosum is the major axon tract that connects and integrates neural activity between the two cerebral hemispheres. Although ∼1:4,000 children are born with developmental absence of the corpus callosum, the primary etiology of this condition remains unknown. Here, we demonstrate that midline crossing of callosal axons is dependent upon the prior remodeling and degradation of the intervening interhemispheric fissure. This remodeling event is initiated by astroglia on either side of the interhemispheric fissure, which intercalate with one another and degrade the intervening leptomeninges. Callosal axons then preferentially extend over these specialized astroglial cells to cross the midline. A key regulatory step in interhemispheric remodeling is the differentiation of these astroglia from radial glia, which is initiated by Fgf8 signaling to downstream Nfi transcription factors. Crucially, our findings from human neuroimaging studies reveal that developmental defects in interhemispheric remodeling are likely to be a primary etiology underlying human callosal agenesis.


Asunto(s)
Astrocitos/metabolismo , Cerebro/embriología , Cuerpo Calloso/embriología , Organogénesis , Agenesia del Cuerpo Calloso/genética , Agenesia del Cuerpo Calloso/patología , Animales , Axones/metabolismo , Diferenciación Celular , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Humanos , Ratones , Fenotipo , Transducción de Señal , Factores de Transcripción/metabolismo
18.
Oncotarget ; 7(20): 29306-20, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27083054

RESUMEN

Glioblastoma (GBM) is an essentially incurable and rapidly fatal cancer, with few markers predicting a favourable prognosis. Here we report that the transcription factor NFIB is associated with significantly improved survival in GBM. NFIB expression correlates inversely with astrocytoma grade and is lowest in mesenchymal GBM. Ectopic expression of NFIB in low-passage, patient-derived classical and mesenchymal subtype GBM cells inhibits tumourigenesis. Ectopic NFIB expression activated phospho-STAT3 signalling only in classical and mesenchymal GBM cells, suggesting a mechanism through which NFIB may exert its context-dependent tumour suppressor activity. Finally, NFIB expression can be induced in GBM cells by drug treatment with beneficial effects.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/patología , Glioblastoma/patología , Factores de Transcripción NFI/metabolismo , Animales , Línea Celular Tumoral , Genes Supresores de Tumor/fisiología , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Supresoras de Tumor/metabolismo
19.
Development ; 142(21): 3746-57, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26534986

RESUMEN

Transcription factors act during cortical development as master regulatory genes that specify cortical arealization and cellular identities. Although numerous transcription factors have been identified as being crucial for cortical development, little is known about their downstream targets and how they mediate the emergence of specific neuronal connections via selective axon guidance. The EMX transcription factors are essential for early patterning of the cerebral cortex, but whether EMX1 mediates interhemispheric connectivity by controlling corpus callosum formation remains unclear. Here, we demonstrate that in mice on the C57Bl/6 background EMX1 plays an essential role in the midline crossing of an axonal subpopulation of the corpus callosum derived from the anterior cingulate cortex. In the absence of EMX1, cingulate axons display reduced expression of the axon guidance receptor NRP1 and form aberrant axonal bundles within the rostral corpus callosum. EMX1 also functions as a transcriptional activator of Nrp1 expression in vitro, and overexpression of this protein in Emx1 knockout mice rescues the midline-crossing phenotype. These findings reveal a novel role for the EMX1 transcription factor in establishing cortical connectivity by regulating the interhemispheric wiring of a subpopulation of neurons within the mouse anterior cingulate cortex.


Asunto(s)
Giro del Cíngulo/metabolismo , Proteínas de Homeodominio/metabolismo , Neuropilina-1/metabolismo , Factores de Transcripción/metabolismo , Agenesia del Cuerpo Calloso/embriología , Agenesia del Cuerpo Calloso/genética , Animales , Axones/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Semaforinas/metabolismo
20.
Stem Cell Reports ; 5(3): 365-77, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26212661

RESUMEN

Mammalian neural stem/progenitor cells (NSPCs) sequentially generate neurons and glia during CNS development. Here we identified miRNA-153 (miR-153) as a modulator of the temporal regulation of NSPC differentiation. Overexpression (OE) of miR-153 delayed the onset of astrogliogenesis and maintained NSPCs in an undifferentiated state in vitro and in the developing cortex. The transcription factors nuclear factor I (NFI) A and B, essential regulators of the initiation of gliogenesis, were found to be targets of miR-153. Inhibition of miR-153 in early neurogenic NSPCs induced precocious gliogenesis, whereas NFIA/B overexpression rescued the anti-gliogenic phenotypes induced by miR-153 OE. Our results indicate that miR-mediated fine control of NFIA/B expression is important in the molecular networks that regulate the acquisition of gliogenic competence by NSPCs in the developing CNS.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/metabolismo , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Animales , Corteza Cerebral/citología , Ratones , MicroARNs/genética , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Neuroglía/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA