Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Dev Biol ; 407(1): 68-74, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26277216

RESUMEN

Gene-targeted knockout technologies are invaluable tools for understanding the functions of genes in vivo. CRISPR/Cas9 system of RNA-guided genome editing is revolutionizing genetics research in a wide spectrum of organisms. Here, we combined CRISPR with in vivo electroporation in the chicken embryo to efficiently target the transcription factor PAX7 in tissues of the developing embryo. This approach generated mosaic genetic mutations within a wild-type cellular background. This series of proof-of-principle experiments indicate that in vivo CRISPR-mediated cell genome engineering is an effective method to achieve gene loss-of-function in the tissues of the chicken embryo and it completes the growing genetic toolbox to study the molecular mechanisms regulating development in this important animal model.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/fisiología , Ingeniería Genética , Animales , Secuencia de Bases , Embrión de Pollo , Pollos , Electroporación , Genoma , Datos de Secuencia Molecular , Factor de Transcripción PAX7/fisiología
2.
Hum Mol Genet ; 23(19): 5147-58, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24833723

RESUMEN

The microtubule cytoskeleton is critical for the generation and maturation of neurons in the developing mammalian nervous system. We have previously shown that mutations in the ß-tubulin gene TUBB5 cause microcephaly with structural brain abnormalities in humans. While it is known that TUBB5 is necessary for the proper generation and migration of neurons, little is understood of the role it plays in neuronal differentiation and connectivity. Here, we report that perturbations to TUBB5 disrupt the morphology of cortical neurons, their neuronal complexity, axonal outgrowth, as well as the density and shape of dendritic spines in the postnatal murine cortex. The features we describe are consistent with defects in synaptic signaling. Cellular-based assays have revealed that TUBB5 substitutions have the capacity to alter the dynamic properties and polymerization rates of the microtubule cytoskeleton. Together, our studies show that TUBB5 is essential for neuronal differentiation and dendritic spine formation in vivo, providing insight into the underlying cellular pathology associated with TUBB5 disease states.


Asunto(s)
Diferenciación Celular/genética , Corteza Cerebral/metabolismo , Espinas Dendríticas/metabolismo , Mutación , Neuronas/citología , Neuronas/metabolismo , Tubulina (Proteína)/genética , Animales , Axones/metabolismo , Corteza Cerebral/embriología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Ratones , Microtúbulos/química , Microtúbulos/metabolismo , Neuronas/patología , Multimerización de Proteína , Interferencia de ARN
3.
Cereb Cortex ; 25(3): 806-16, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24084125

RESUMEN

The zinc finger transcription factor RP58 (also known as ZNF238) regulates neurogenesis of the mouse neocortex and cerebellum (Okado et al. 2009; Xiang et al. 2011; Baubet et al. 2012; Ohtaka-Maruyama et al. 2013), but its mechanism of action remains unclear. In this study, we report a cell-autonomous function for RP58 during the differentiation of embryonic cortical projection neurons via its activities as a transcriptional repressor. Disruption of RP58 expression alters the differentiation of immature neurons and impairs their migration and positioning within the mouse cerebral cortex. Loss of RP58 within the embryonic cortex also leads to elevated mRNA for Rnd2, a member of the Rnd family of atypical RhoA-like GTPase proteins important for cortical neuron migration (Heng et al. 2008). Mechanistically, RP58 represses transcription of Rnd2 via binding to a 3'-regulatory enhancer in a sequence-specific fashion. Using reporter assays, we found that RP58 repression of Rnd2 is competed by proneural basic helix-loop-helix transcriptional activators. Finally, our rescue experiments revealed that negative regulation of Rnd2 by RP58 was important for cortical cell migration in vivo. Taken together, these studies demonstrate that RP58 is a key player in the transcriptional control of cell migration in the developing cerebral cortex.


Asunto(s)
Movimiento Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Neuronas/metabolismo , Proteínas Represoras/genética , Proteínas de Unión al GTP rho/genética , Animales , Proliferación Celular/genética , Corteza Cerebral/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados
4.
J Cell Sci ; 125(Pt 21): 5096-109, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22899712

RESUMEN

The impact of aberrant centrosomes and/or spindles on asymmetric cell division in embryonic development indicates the tight regulation of bipolar spindle formation and positioning that is required for mitotic progression and cell fate determination. WD40-repeat protein 62 (WDR62) was recently identified as a spindle pole protein linked to the neurodevelopmental defect of microcephaly but its roles in mitosis have not been defined. We report here that the in utero electroporation of neuroprogenitor cells with WDR62 siRNAs induced their cell cycle exit and reduced their proliferative capacity. In cultured cells, we demonstrated cell-cycle-dependent accumulation of WDR62 at the spindle pole during mitotic entry that persisted until metaphase-anaphase transition. Utilizing siRNA depletion, we revealed WDR62 function in stabilizing the mitotic spindle specifically during metaphase. WDR62 loss resulted in spindle orientation defects, decreased the integrity of centrosomes displaced from the spindle pole and delayed mitotic progression. Additionally, we revealed JNK phosphorylation of WDR62 is required for maintaining metaphase spindle organization during mitosis. Our study provides the first functional characterization of WDR62 and has revealed requirements for JNK/WDR62 signaling in mitotic spindle regulation that may be involved in coordinating neurogenesis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metafase , Proteínas Asociadas a Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Huso Acromático/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Centrosoma/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Microcefalia , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Fosforilación , Profase , Transporte de Proteínas , ARN Interferente Pequeño/genética
5.
Eur J Neurosci ; 37(10): 1584-93, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23451919

RESUMEN

During brain development, many factors influence the assembly and final positioning of cortical neurons, and this process is essential for proper circuit formation and normal brain function. Among many important extrinsic factors that guide the maturation of embryonic cortical neurons, the secreted neurotransmitter GABA has been proposed to influence both their migratory behaviour and their terminal differentiation. The full extent of the short-term and long-term changes in brain patterning and function caused by modulators of the GABA system is not known. In this study, we specifically investigated whether diazepam, a commonly used benzodiazepine that modulates the GABAA receptor, alters neuronal positioning in vivo, and whether this can lead to lasting effects on brain function. We found that fetal exposure to diazepam did not change cell positioning within the embryonic day (E)14.5 mouse cerebral cortex, but significantly altered neuron positioning within the E18.5 cortex. In adult mice, diazepam treatment affected the distribution of cortical interneurons that express parvalbumin or calretinin, and also led to a decrease in the numbers of calretinin-expressing interneurons. In addition, we observed that neonatal exposure to diazepam altered the sensitivity of mice to a proconvulsant challenge. Therefore, exposure of the fetal brain to benzodiazepines has consequences for the positioning of neurons and cortical network excitability.


Asunto(s)
Anticonvulsivantes/farmacología , Corteza Cerebral/efectos de los fármacos , Diazepam/farmacología , Moduladores del GABA/farmacología , Interneuronas/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Anticonvulsivantes/uso terapéutico , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/fisiopatología , Diazepam/uso terapéutico , Femenino , Moduladores del GABA/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Embarazo , Convulsiones/dietoterapia
6.
J Physiol ; 589(Pt 2): 325-39, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21098001

RESUMEN

In the present study the relationship between tissue damage and changed electro-physiological properties of Dogiel type II myenteric neurons within the first 24 hours after induction of inflammation with trinitrobenzene sulfonate (TNBS) in the guinea-pig ileum was investigated. Treatment with TNBS causes damage to the mucosa, inflammatory responses in the mucosa and enteric ganglia and changes in myenteric neuron properties. Thus we hypothesise that the physiological changes in the myenteric neurons could be due to damage to their mucosal processes or inflammation in the vicinity of cell bodies or the processes. We found an association between hyperexcitability of myenteric Dogiel type II neurons and damage to the mucosa and its innervation at 3 and 24 h, times when there was also an inflammatory reaction. The lack of hyperexcitability in neurons from control tissues in which axons projecting to the mucosa were severed suggests that inflammation may be an important contributing factor to the neuronal hyperexcitability at the acute stage of inflammation. Despite mucosal repair and re-innervation of the mucosa before 7 days after induction of inflammation, neuronal hyperexcitability persists. Although the mechanisms underlying neuronal hyperexcitability at the acute stage of inflammation might be different from those underlying long-term changes in the absence of active inflammation in the ganglia, the persistent changes in neuronal excitability may contribute to post-inflammatory gut dysfunctions.


Asunto(s)
Íleon/inervación , Inflamación/patología , Mucosa Intestinal/inervación , Plexo Mientérico/patología , Neuronas/patología , Animales , Calbindinas , Recuento de Células , Estimulación Eléctrica , Electrofisiología , Femenino , Cobayas , Inmunohistoquímica , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Potenciales de la Membrana/fisiología , Microscopía Confocal , Plexo Mientérico/metabolismo , Plexo Mientérico/fisiopatología , Neuronas/fisiología , Proteína G de Unión al Calcio S100/metabolismo , Ácido Trinitrobencenosulfónico
7.
Cell Tissue Res ; 334(2): 147-61, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18855018

RESUMEN

The definition of the nerve cell types of the myenteric plexus of the mouse small intestine has become important, as more researchers turn to the use of mice with genetic mutations to analyze roles of specific genes and their products in enteric nervous system function and to investigate animal models of disease. We have used a suite of antibodies to define neurons by their shapes, sizes, and neurochemistry in the myenteric plexus. Anti-Hu antibodies were used to reveal all nerve cells, and the major subpopulations were defined in relation to the Hu-positive neurons. Morphological Type II neurons, revealed by anti-neurofilament and anti-calcitonin gene-related peptide antibodies, represented 26% of neurons. The axons of the Type II neurons projected through the circular muscle and submucosa to the mucosa. The cell bodies were immunoreactive for choline acetyltransferase (ChAT), and their terminals were immunoreactive for vesicular acetylcholine transporter (VAChT). Nitric oxide synthase (NOS) occurred in 29% of nerve cells. Most were also immunoreactive for vasoactive intestinal peptide, but they were not tachykinin (TK)-immunoreactive, and only 10% were ChAT-immunoreactive. Numerous NOS terminals occurred in the circular muscle. We deduced that 90% of NOS neurons were inhibitory motor neurons to the muscle (26% of all neurons) and 10% (3% of all neurons) were interneurons. Calretinin immunoreactivity was found in a high proportion of neurons (52%). Many of these had TK immunoreactivity. Small calretinin neurons were identified as excitatory neurons to the longitudinal muscle (about 20% of neurons, with ChAT/calretinin/+/- TK chemical coding). Excitatory neurons to the circular muscle (about 10% of neurons) had the same coding. Calretinin immunoreactivity also occurred in a proportion of Type II neurons. Thus, over 90% of neurons in the myenteric plexus of the mouse small intestine can be currently identified by their neurochemistry and shape.


Asunto(s)
Axones/metabolismo , Intestino Delgado/inervación , Plexo Mientérico/ultraestructura , Neuronas/clasificación , Neuronas/ultraestructura , Plexo Submucoso/ultraestructura , Animales , Calbindina 2 , Calbindinas , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colina O-Acetiltransferasa/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Plexo Mientérico/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Somatostatina/metabolismo , Plexo Submucoso/metabolismo , Taquicininas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
8.
Neural Dev ; 13(1): 1, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29325591

RESUMEN

CORRECTION: After publication of the original article [1] it was realised that there were errors in figures 2a,b,f,g, which arose as a result of preparing figures from data collected and analysed at the same time as the work reported in [2] (Supplementary Figure 1 of [2]). An updated Fig. 2 is included with this Correction.

9.
Neural Dev ; 12(1): 8, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506232

RESUMEN

BACKGROUND: During the development of the mammalian cerebral cortex, newborn postmitotic projection neurons are born from local neural stem cells and must undergo radial migration so as to position themselves appropriately to form functional neural circuits. The zinc finger transcriptional repressor Rp58 (also known as Znf238 or Zbtb18) is critical for coordinating corticogenesis, but its underlying molecular mechanism remains to be better characterised. FINDINGS: Here, we demonstrate that the co-expression of Rp58 and the cyclin dependent kinase inhibitor (CDKI) p27kip1 is important for E14.5-born cortical neurons to coordinate cell cycle exit and initiate their radial migration. Notably, we find that the impaired radial positioning of Rp58-deficient cortical neurons within the embryonic (E17.5) mouse cortex, as well as their multipolar to bipolar transition from the intermediate zone to the cortical plate can be restored by forced expression of p27kip1 in concert with suppression of Rnd2, a downstream target gene of Rp58. Furthermore, the restorative effects of p27kip1 and Rnd2 abrogation are reminiscent of suppressing RhoA signalling in Rp58-deficient cells. CONCLUSIONS: Our findings demonstrate functional interplay between a transcriptional regulator and a CDKI to mediate neuroprogenitor cell cycle exit, as well as to promote radial migration through a molecular mechanism consistent with suppression of RhoA signalling.


Asunto(s)
Ciclo Celular , Movimiento Celular , Corteza Cerebral/embriología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Neuronas/fisiología , Proteínas Represoras/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo
10.
Neural Dev ; 11: 7, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26969432

RESUMEN

BACKGROUND: The development of neural circuits within the embryonic cerebral cortex relies on the timely production of neurons, their positioning within the embryonic cerebral cortex as well as their terminal differentiation and dendritic spine connectivity. The RhoA GTPases Rnd2 and Rnd3 are important for neurogenesis and cell migration within the embryonic cortex (Nat Commun 4:1635, 2013), and we recently identified the BTB/POZ domain-containing Adaptor for Cul3-mediated RhoA Degradation family member Bacurd2 (also known as Tnfaip1) as an interacting partner to Rnd2 for the migration of embryonic mouse cortical neurons (Neural Dev 10:9, 2015). FINDINGS: We have extended this work and report that Bacurd1/Kctd13 and Bacurd2/Tnfaip1 are interacting partners to Rnd2 and Rnd3 in vitro. Given that these genes are expressed during cortical development, we performed a series of in utero electroporation studies in mice and found that disruptions to Bacurd1/Kctd13 or Bacurd2/Tnfaip1 expression impair the long-term positioning of E14.5-born cortical neurons within the postnatal (P17) mouse cerebral cortex. We also find that forced expression of Bacurd1/Kctd13 and Bacurd2/Tnfaip1 alters the branching and dendritic spine properties of layer II/III projection neurons. CONCLUSIONS: We identify Bacurd1/Kctd13 and Bacurd2/Tnfaip1 as interacting partners to Rnd proteins which influence the development of cortical neurons. Their neurodevelopmental functions are likely to be relevant to human brain development and disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Corteza Cerebral/embriología , Espinas Dendríticas/fisiología , Proteínas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/fisiología , Movimiento Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Proteínas/fisiología , Complejos de Ubiquitina-Proteína Ligasa , Proteínas de Unión al GTP rho/fisiología
11.
Mol Autism ; 7: 22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27042284

RESUMEN

BACKGROUND: BMP/RA-inducible neural-specific protein 1 (Brinp1) is highly conserved in vertebrates, and continuously expressed in the neocortex, hippocampus, olfactory bulb and cerebellum from mid-embryonic development through to adulthood. METHODS: Brinp1 knock-out (Brinp1(-/-)) mice were generated by Cre-recombinase-mediated removal of the third exon of Brinp1. Knock-out mice were characterised by behavioural phenotyping, immunohistochemistry and expression analysis of the developing and adult brain. RESULTS: Absence of Brinp1 during development results in a behavioural phenotype resembling autism spectrum disorder (ASD), in which knock-out mice show reduced sociability and changes in vocalisation capacity. In addition, Brinp1(-/-) mice exhibit hyper-locomotor activity, have impaired short-term memory, and exhibit poor reproductive success. Brinp1(-/-) mice show increased density of parvalbumin-expressing interneurons in the adult mouse brain. Brinp1(-/-) mice do not show signs of altered neural precursor proliferation or increased apoptosis during late embryonic brain development. The expression of the related neuronal migration genes Astn1 and Astn2 is increased in the brains of Brinp1(-/-) mice, suggesting that they may ameliorate the effects of Brinp1 loss. CONCLUSIONS: Brinp1 plays an important role in normal brain development and function by influencing neuronal distribution within the cortex. The increased cortical PV-positive interneuron density and altered behaviour of Brinp1(-/-) mice resemble features of a subset of human neurological disorders; namely autism spectrum disorder (ASD) and the hyperactivity aspect of attention deficit hyperactivity disorder (ADHD).


Asunto(s)
Trastorno del Espectro Autista/patología , Proteínas del Tejido Nervioso/genética , Animales , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno por Déficit de Atención con Hiperactividad/patología , Trastorno del Espectro Autista/metabolismo , Conducta Animal , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Ciclo Celular , Modelos Animales de Enfermedad , Femenino , Genotipo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Interneuronas/metabolismo , Masculino , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Parvalbúminas/genética , Parvalbúminas/metabolismo , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Vocalización Animal
12.
Sci Rep ; 6: 29514, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27404227

RESUMEN

Copy number variations to chromosome 21 (HSA21) cause intellectual disability and Down Syndrome, but our understanding of the HSA21 genetic factors which contribute to fetal brain development remains incomplete. Here, we focussed on the neurodevelopmental functions for EURL (also known as C21ORF91, Refseq Gene ID:54149), a protein-coding gene at the centromeric boundary of the Down Syndrome Critical Region (DSCR) of HSA21. We report that EURL is expressed during human and mouse cerebral cortex development, and we report that alterations to EURL mRNA levels within the human brain underlie Down Syndrome. Our gene perturbation studies in mice demonstrate that disruptions to Eurl impair progenitor proliferation and neuronal differentiation. Also, we find that disruptions to Eurl impair the long-term positioning and dendritic spine densities of cortical projection neurons. We provide evidence that EURL interacts with the coiled-coil domain-containing protein CCDC85B so as to modulate ß-catenin levels in cells. Further, we utilised a fluorescent reporter (8xTOPFLASHd2EGFP) to demonstrate that disruptions to Eurl alter ß-catenin signalling in vitro as well as in vivo. Together, these studies highlight EURL as an important new player in neuronal development that is likely to impact on the neuropathogenesis of HSA21-related disorders including Down Syndrome.


Asunto(s)
Corteza Cerebral/embriología , Cromosomas Humanos Par 21/genética , Síndrome de Down/genética , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Variaciones en el Número de Copia de ADN/genética , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Síndrome de Down/patología , Humanos , Discapacidad Intelectual/genética , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/metabolismo , beta Catenina/metabolismo
13.
Cell Rep ; 15(10): 2251-2265, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27239039

RESUMEN

Disruptions to neuronal mRNA translation are hypothesized to underlie human neurodevelopmental syndromes. Notably, the mRNA translation re-initiation factor DENR is a regulator of eukaryotic translation and cell growth, but its mammalian functions are unknown. Here, we report that Denr influences the migration of murine cerebral cortical neurons in vivo with its binding partner Mcts1, whereas perturbations to Denr impair the long-term positioning, dendritic arborization, and dendritic spine characteristics of postnatal projection neurons. We characterized de novo missense mutations in DENR (p.C37Y and p.P121L) detected in two unrelated human subjects diagnosed with brain developmental disorder to find that each variant impairs the function of DENR in mRNA translation re-initiation and disrupts the migration and terminal branching of cortical neurons in different ways. Thus, our findings link human brain disorders to impaired mRNA translation re-initiation through perturbations in DENR (OMIM: 604550) function in neurons.


Asunto(s)
Factores Eucarióticos de Iniciación/genética , Mutación/genética , Enfermedades del Sistema Nervioso/congénito , Enfermedades del Sistema Nervioso/genética , Neurogénesis/genética , Neuronas/metabolismo , Iniciación de la Cadena Peptídica Traduccional/genética , Animales , Diferenciación Celular , Movimiento Celular , Corteza Cerebral/embriología , Corteza Cerebral/patología , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Neural Dev ; 10: 9, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25888806

RESUMEN

BACKGROUND: During fetal brain development in mammals, newborn neurons undergo cell migration to reach their appropriate positions and form functional circuits. We previously reported that the atypical RhoA GTPase Rnd2 promotes the radial migration of mouse cerebral cortical neurons (Nature 455(7209):114-8, 2008; Neuron 69(6):1069-84, 2011), but its downstream signalling pathway is not well understood. RESULTS: We have identified BTB-domain containing adaptor for Cul3-mediated RhoA degradation 2 (Bacurd2) as a novel interacting partner to Rnd2, which promotes radial migration within the developing cerebral cortex. We find that Bacurd2 binds Rnd2 at its C-terminus, and this interaction is critical to its cell migration function. We show that forced expression or knockdown of Bacurd2 impairs neuronal migration within the embryonic cortex and alters the morphology of immature neurons. Our in vivo cellular analysis reveals that Bacurd2 influences the multipolar-to-bipolar transition of radially migrating neurons in a cell autonomous fashion. When we addressed the potential signalling relationship between Bacurd2 and Rnd2 using a Bacurd2-Rnd2 chimeric construct, our results suggest that Bacurd2 and Rnd2 could interact to promote radial migration within the embryonic cortex. CONCLUSIONS: Our studies demonstrate that Bacurd2 is a novel player in neuronal development and influences radial migration within the embryonic cerebral cortex.


Asunto(s)
Corteza Cerebral/embriología , Red Nerviosa/embriología , Proteínas de Unión al GTP rho/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Movimiento Celular , Corteza Cerebral/citología , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección , Técnicas del Sistema de Dos Híbridos
15.
Cell Rep ; 2(6): 1554-62, 2012 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-23246003

RESUMEN

The formation of the mammalian cortex requires the generation, migration, and differentiation of neurons. The vital role that the microtubule cytoskeleton plays in these cellular processes is reflected by the discovery that mutations in various tubulin isotypes cause different neurodevelopmental diseases, including lissencephaly (TUBA1A), polymicrogyria (TUBA1A, TUBB2B, TUBB3), and an ocular motility disorder (TUBB3). Here, we show that Tubb5 is expressed in neurogenic progenitors in the mouse and that its depletion in vivo perturbs the cell cycle of progenitors and alters the position of migrating neurons. We report the occurrence of three microcephalic patients with structural brain abnormalities harboring de novo mutations in TUBB5 (M299V, V353I, and E401K). These mutant proteins, which affect the chaperone-dependent assembly of tubulin heterodimers in different ways, disrupt neurogenic division and/or migration in vivo. Our results provide insight into the functional repertoire of the tubulin gene family, specifically implicating TUBB5 in embryonic neurogenesis and microcephaly.


Asunto(s)
Encéfalo/anomalías , Encéfalo/metabolismo , Microcefalia/metabolismo , Mutación Missense , Células-Madre Neurales/metabolismo , Tubulina (Proteína)/metabolismo , Sustitución de Aminoácidos , Animales , Encéfalo/embriología , Encéfalo/patología , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Microcefalia/embriología , Microcefalia/genética , Microcefalia/patología , Células-Madre Neurales/patología , Neurogénesis/genética , Tubulina (Proteína)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA