Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Neurobiol Dis ; 198: 106558, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38852754

RESUMEN

Periventricular nodular heterotopia (PNH), the most common brain malformation diagnosed in adulthood, is characterized by the presence of neuronal nodules along the ventricular walls. PNH is mainly associated with mutations in the FLNA gene - encoding an actin-binding protein - and patients often develop epilepsy. However, the molecular mechanisms underlying the neuronal failure still remain elusive. It has been hypothesized that dysfunctional cortical circuitry, rather than ectopic neurons, may explain the clinical manifestations. To address this issue, we depleted FLNA from cortical pyramidal neurons of a conditional Flnaflox/flox mice by timed in utero electroporation of Cre recombinase. We found that FLNA regulates dendritogenesis and spinogenesis thus promoting an appropriate excitatory/inhibitory inputs balance. We demonstrated that FLNA modulates RAC1 and cofilin activity through its interaction with the Rho-GTPase Activating Protein 24 (ARHGAP24). Collectively, we disclose an uncharacterized role of FLNA and provide strong support for neural circuit dysfunction being a consequence of FLNA mutations.


Asunto(s)
Corteza Cerebral , Filaminas , Proteína de Unión al GTP rac1 , Animales , Ratones , Factores Despolimerizantes de la Actina/metabolismo , Corteza Cerebral/metabolismo , Filaminas/metabolismo , Filaminas/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/genética , Ratones Transgénicos , Neurogénesis/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Neuropéptidos/genética , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/metabolismo , Heterotopia Nodular Periventricular/patología , Células Piramidales/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética
2.
J Physiol ; 600(10): 2429-2460, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35389519

RESUMEN

De novo missense variants in the KCNQ2 gene encoding the Kv7.2 subunit of voltage-gated potassium Kv7/M channels are the main cause of developmental and epileptic encephalopathy with neonatal onset. Although seizures usually resolve during development, cognitive/motor deficits persist. To gain a better understanding of the cellular mechanisms underlying network dysfunction and their progression over time, we investigated in vivo, using local field potential recordings of freely moving animals, and ex vivo in layers II/III and V of motor cortical slices, using patch-clamp recordings, the electrophysiological properties of pyramidal cells from a heterozygous knock-in mouse model carrying the Kv7.2 p.T274M pathogenic variant during neonatal, postweaning and juvenile developmental stages. We found that knock-in mice displayed spontaneous seizures preferentially at postweaning rather than at juvenile stages. At the cellular level, the variant led to a reduction in M ​​current density/conductance and to neuronal hyperexcitability. These alterations were observed during the neonatal period in pyramidal cells of layers II/III and during the postweaning stage in pyramidal cells of layer V. Moreover, there was an increase in the frequency of spontaneous network-driven events mediated by GABA receptors, suggesting that the excitability of interneurons was also increased. However, all these alterations were no longer observed in layers II/III and V of juvenile mice. Thus, our data indicate that the action of the variant is regulated developmentally. This raises the possibility that the age-related seizure remission observed in KCNQ2-related developmental and epileptic encephalopathy patients results from a time-limited alteration of Kv7 channel activity and neuronal excitability. KEY POINTS: The electrophysiological impact of the pathogenic c.821C>T mutation of the KCNQ2 gene (p.T274M variant in Kv7.2 subunit) related to developmental and epileptic encephalopathy has been analysed both in vivo and ex vivo in layers II/III and V of motor cortical slices from a knock-in mouse model during development at neonatal, postweaning and juvenile stages. M current density and conductance are decreased and the excitability of layer II/III pyramidal cells is increased in slices from neonatal and postweaning knock-in mice but not from juvenile knock-in mice. M current and excitability of layer V pyramidal cells are impacted in knock-in mice only at the postweaning stage. Spontaneous GABAergic network-driven events can be recorded until the postweaning stage, and their frequency is increased in layers II/III of the knock-in mice. Knock-in mice display spontaneous seizures preferentially at postweaning rather than at juvenile stages.


Asunto(s)
Encefalopatías , Canal de Potasio KCNQ2 , Convulsiones , Animales , Modelos Animales de Enfermedad , Humanos , Canal de Potasio KCNQ2/genética , Ratones , Proteínas del Tejido Nervioso , Células Piramidales
3.
Mol Psychiatry ; 26(11): 6125-6148, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34188164

RESUMEN

While the transcription factor NEUROD2 has recently been associated with epilepsy, its precise role during nervous system development remains unclear. Using a multi-scale approach, we set out to understand how Neurod2 deletion affects the development of the cerebral cortex in mice. In Neurod2 KO embryos, cortical projection neurons over-migrated, thereby altering the final size and position of layers. In juvenile and adults, spine density and turnover were dysregulated in apical but not basal compartments in layer 5 neurons. Patch-clamp recordings in layer 5 neurons of juvenile mice revealed increased intrinsic excitability. Bulk RNA sequencing showed dysregulated expression of many genes associated with neuronal excitability and synaptic function, whose human orthologs were strongly associated with autism spectrum disorders (ASD). At the behavior level, Neurod2 KO mice displayed social interaction deficits, stereotypies, hyperactivity, and occasionally spontaneous seizures. Mice heterozygous for Neurod2 had similar defects, indicating that Neurod2 is haploinsufficient. Finally, specific deletion of Neurod2 in forebrain excitatory neurons recapitulated cellular and behavioral phenotypes found in constitutive KO mice, revealing the region-specific contribution of dysfunctional Neurod2 in symptoms. Informed by these neurobehavioral features in mouse mutants, we identified eleven patients from eight families with a neurodevelopmental disorder including intellectual disability and ASD associated with NEUROD2 pathogenic mutations. Our findings demonstrate crucial roles for Neurod2 in neocortical development, whose alterations can cause neurodevelopmental disorders including intellectual disability and ASD.


Asunto(s)
Trastorno Autístico , Neuropéptidos , Animales , Trastorno Autístico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Corteza Cerebral/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Neuropéptidos/metabolismo , Prosencéfalo/metabolismo , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(6): 2337-42, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24469796

RESUMEN

Alterations in the formation of brain networks are associated with several neurodevelopmental disorders. Mutations in TBC1 domain family member 24 (TBC1D24) are responsible for syndromes that combine cortical malformations, intellectual disability, and epilepsy, but the function of TBC1D24 in the brain remains unknown. We report here that in utero TBC1D24 knockdown in the rat developing neocortex affects the multipolar-bipolar transition of neurons leading to delayed radial migration. Furthermore, we find that TBC1D24-knockdown neurons display an abnormal maturation and retain immature morphofunctional properties. TBC1D24 interacts with ADP ribosylation factor (ARF)6, a small GTPase crucial for membrane trafficking. We show that in vivo, overexpression of the dominant-negative form of ARF6 rescues the neuronal migration and dendritic outgrowth defects induced by TBC1D24 knockdown, suggesting that TBC1D24 prevents ARF6 activation. Overall, our findings demonstrate an essential role of TBC1D24 in neuronal migration and maturation and highlight the physiological relevance of the ARF6-dependent membrane-trafficking pathway in brain development.


Asunto(s)
Factores de Ribosilacion-ADP/fisiología , Proteínas Portadoras/fisiología , Movimiento Celular/fisiología , Neuronas/citología , Factor 6 de Ribosilación del ADP , Animales , Encéfalo/fisiología , Proteínas Portadoras/genética , Células Cultivadas , Dendritas/fisiología , Proteínas Activadoras de GTPasa , Técnicas de Silenciamiento del Gen , Ácido Glutámico/metabolismo , Proteínas de la Membrana , Proteínas del Tejido Nervioso , Ratas , Sinapsis/metabolismo
6.
Hum Mol Genet ; 21(5): 1004-17, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22076441

RESUMEN

Periventricular nodular heterotopia (PH) is a human brain malformation caused by defective neuronal migration that results in ectopic neuronal nodules lining the lateral ventricles beneath a normal appearing cortex. Most affected patients have seizures and their cognitive level varies from normal to severely impaired. Mutations in the Filamin-A (or FLNA) gene are the main cause of PH, but the underlying pathological mechanism remains unknown. Although two FlnA knockout mouse strains have been generated, none of them showed the presence of ectopic nodules. To recapitulate the loss of FlnA function in the developing rat brain, we used an in utero RNA interference-mediated knockdown approach and successfully reproduced a PH phenotype in rats comparable with that observed in human patients. In FlnA-knockdown rats, we report that PH results from a disruption of the polarized radial glial scaffold in the ventricular zone altering progression of neural progenitors through the cell cycle and impairing migration of neurons into the cortical plate. Similar alterations of radial glia are observed in human PH brains of a 35-week fetus and a 3-month-old child, harboring distinct FLNA mutations not previously reported. Finally, juvenile FlnA-knockdown rats are highly susceptible to seizures, confirming the reliability of this novel animal model of PH. Our findings suggest that the disorganization of radial glia is the leading cause of PH pathogenesis associated with FLNA mutations. Rattus norvegicus FlnA mRNA (GenBank accession number FJ416060).


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Neuroglía/fisiología , Heterotopia Nodular Periventricular/metabolismo , Heterotopia Nodular Periventricular/patología , Animales , Movimiento Celular , Proliferación Celular , Corteza Cerebral/embriología , Corteza Cerebral/patología , Ventrículos Cerebrales/patología , Proteínas Contráctiles/genética , Modelos Animales de Enfermedad , Femenino , Filaminas , Humanos , Lactante , Proteínas de Microfilamentos/genética , Datos de Secuencia Molecular , Neocórtex/embriología , Neocórtex/metabolismo , Neocórtex/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Neuroglía/metabolismo , Neuroglía/ultraestructura , Neuronas/fisiología , Interferencia de ARN , Ratas , Convulsiones/etiología
7.
Brain ; 136(Pt 11): 3378-94, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24056535

RESUMEN

Periventricular nodular heterotopia is caused by defective neuronal migration that results in heterotopic neuronal nodules lining the lateral ventricles. Mutations in filamin A (FLNA) or ADP-ribosylation factor guanine nucleotide-exchange factor 2 (ARFGEF2) cause periventricular nodular heterotopia, but most patients with this malformation do not have a known aetiology. Using comparative genomic hybridization, we identified 12 patients with developmental brain abnormalities, variably combining periventricular nodular heterotopia, corpus callosum dysgenesis, colpocephaly, cerebellar hypoplasia and polymicrogyria, harbouring a common 1.2 Mb minimal critical deletion in 6q27. These anatomic features were mainly associated with epilepsy, ataxia and cognitive impairment. Using whole exome sequencing in 14 patients with isolated periventricular nodular heterotopia but no copy number variants, we identified one patient with periventricular nodular heterotopia, developmental delay and epilepsy and a de novo missense mutation in the chromosome 6 open reading frame 70 (C6orf70) gene, mapping in the minimal critical deleted region. Using immunohistochemistry and western blots, we demonstrated that in human cell lines, C6orf70 shows primarily a cytoplasmic vesicular puncta-like distribution and that the mutation affects its stability and subcellular distribution. We also performed in utero silencing of C6orf70 and of Phf10 and Dll1, the two additional genes mapping in the 6q27 minimal critical deleted region that are expressed in human and rodent brain. Silencing of C6orf70 in the developing rat neocortex produced periventricular nodular heterotopia that was rescued by concomitant expression of wild-type human C6orf70 protein. Silencing of the contiguous Phf10 or Dll1 genes only produced slightly delayed migration but not periventricular nodular heterotopia. The complex brain phenotype observed in the 6q terminal deletion syndrome likely results from the combined haploinsufficiency of contiguous genes mapping to a small 1.2 Mb region. Our data suggest that, of the genes within this minimal critical region, C6orf70 plays a major role in the control of neuronal migration and its haploinsufficiency or mutation causes periventricular nodular heterotopia.


Asunto(s)
Anomalías Múltiples/genética , Encéfalo/anomalías , Malformaciones del Desarrollo Cortical del Grupo II/genética , Heterotopia Nodular Periventricular/genética , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Adolescente , Adulto , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Niño , Deleción Cromosómica , Cromosomas Humanos Par 6/genética , Estudios de Cohortes , Discapacidades del Desarrollo/genética , Epilepsia/genética , Exoma/genética , Femenino , Haploinsuficiencia/genética , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Malformaciones del Desarrollo Cortical del Grupo II/patología , Malformaciones del Desarrollo Cortical del Grupo II/fisiopatología , Mutación/genética , Heterotopia Nodular Periventricular/patología , Heterotopia Nodular Periventricular/fisiopatología , Ratas , Ratas Wistar , Síndrome
8.
Cereb Cortex ; 23(6): 1484-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22628459

RESUMEN

Epileptic encephalopathies comprise a heterogeneous group of severe infantile disorders for which the pathophysiological basis of epilepsy is inaccurately clarified by genotype-phenotype analysis. Because a deficit of GABA neurons has been found in some of these syndromes, notably in patients with X-linked lissencephaly with abnormal genitalia, epilepsy was suggested to result from an imbalance in GABAergic inhibition, and the notion of "interneuronopathy" was proposed. Here, we studied the impact of a polyalanine expansion of aristaless-related homeobox (ARX) gene, a mutation notably found in West and Ohtahara syndromes. Analysis of Arx((GCG)7/Y) knock-in mice revealed that GABA neuron development is not affected. Moreover, pyramidal cell migration and cortical layering are unaltered in these mice. Interestingly, electrophysiological recordings show that hippocampal pyramidal neurons displayed a frequency of inhibitory postsynaptic currents similar to wild-type (WT) mice. However, these neurons show a dramatic increase in the frequency of excitatory inputs associated with a remodeling of their axonal arborization, suggesting that epilepsy in Arx((GCG)7/Y)mice would result from a glutamate network remodeling. We therefore propose that secondary alterations are instrumental for the development of disease-specific phenotypes and should be considered to explain the phenotypic diversity associated with epileptogenic mutations.


Asunto(s)
Neuronas GABAérgicas/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Glutamatos/metabolismo , Proteínas de Homeodominio/genética , Péptidos/genética , Factores de Transcripción/genética , Ácido gamma-Aminobutírico/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Factores de Edad , Animales , Animales Recién Nacidos , Movimiento Celular/genética , Proteína Doblecortina , Electroporación , Embrión de Mamíferos , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Neuronas GABAérgicas/citología , Glutamato Descarboxilasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , ARN Interferente Pequeño/genética , Estadísticas no Paramétricas , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética , Transfección
9.
Neuropsychopharmacology ; 47(11): 1901-1912, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35396500

RESUMEN

Atypical responses to sensory stimuli are considered as a core aspect and early life marker of autism spectrum disorders (ASD). Although recent findings performed in mouse ASD genetic models report sensory deficits, these were explored exclusively during juvenile or adult period. Whether sensory dysfunctions might be present at the early life stage and rescued by therapeutic strategy are fairly uninvestigated. Here we found that under cool environment neonatal mice lacking the autism-associated gene Magel2 present pup calls hypo-reactivity and are retrieved with delay by their wild-type dam. This neonatal atypical sensory reactivity to cool stimuli was not associated with autonomic thermoregulatory alteration but with a deficit of the oxytocinergic system. Indeed, we show in control neonates that pharmacogenetic inactivation of hypothalamic oxytocin neurons mimicked atypical thermosensory reactivity found in Magel2 mutants. Furthermore, pharmacological intranasal administration of oxytocin to Magel2 neonates was able to rescue both the atypical thermosensory response and the maternal pup retrieval. This preclinical study establishes for the first-time early life impairments in thermosensory integration and suggest a therapeutic potential benefit of intranasal oxytocin treatment on neonatal atypical sensory reactivity for autism.


Asunto(s)
Trastorno Autístico , Hipoestesia , Conducta Materna , Oxitocina , Proteínas , Administración Intranasal , Factores de Edad , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Trastorno del Espectro Autista/complicaciones , Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Fármacos del Sistema Nervioso Central/administración & dosificación , Fármacos del Sistema Nervioso Central/metabolismo , Femenino , Hipoestesia/etiología , Hipoestesia/genética , Hipoestesia/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Conducta Materna/fisiología , Ratones , Oxitocina/administración & dosificación , Oxitocina/metabolismo , Proteínas/genética , Proteínas/metabolismo , Conducta Social
10.
Biochim Biophys Acta ; 1778(3): 614-30, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18005931

RESUMEN

The formation of functional epithelial tissues involves the coordinated action of several protein complexes, which together produce a cell polarity axis and develop cell-cell junctions. During the last decade, the notion of polarity complexes emerged as the result of genetic studies in which a set of genes was discovered first in Caenorhabditis elegans and then in Drosophila melanogaster. In epithelial cells, these complexes are responsible for the development of the apico-basal axis and for the construction and maintenance of apical junctions. In this review, we focus on apical polarity complexes, namely the PAR3/PAR6/aPKC complex and the CRUMBS/PALS1/PATJ complex, which are conserved between species and along with a lateral complex, the SCRIBBLE/DLG/LGL complex, are crucial to the formation of apical junctions such as tight junctions in mammalian epithelial cells. The exact mechanisms underlying their tight junction construction and maintenance activities are poorly understood, and it is proposed to focus in this review on establishing how these apical polarity complexes might regulate epithelial cell morphogenesis and functions. In particular, we will present the latest findings on how these complexes regulate epithelial homeostasis.


Asunto(s)
Polaridad Celular/fisiología , Uniones Intercelulares/fisiología , Proteínas de la Membrana/fisiología , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Biológicos , Estructura Molecular , Complejos Multiproteicos
11.
Front Cell Neurosci ; 12: 55, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29559892

RESUMEN

Congenital cytomegalovirus (CMV) infections represent one leading cause of neurodevelopmental disorders. Recently, we reported on a rat model of CMV infection of the developing brain in utero, characterized by early and prominent infection and alteration of microglia-the brain-resident mononuclear phagocytes. Besides their canonical function against pathogens, microglia are also pivotal to brain development. Here we show that CMV infection of the rat fetal brain recapitulated key postnatal phenotypes of human congenital CMV including increased mortality, sensorimotor impairment reminiscent of cerebral palsy, hearing defects, and epileptic seizures. The possible influence of early microglia alteration on those phenotypes was then questioned by pharmacological targeting of microglia during pregnancy. One single administration of clodronate liposomes in the embryonic brains at the time of CMV injection to deplete microglia, and maternal feeding with doxycyxline throughout pregnancy to modify microglia in the litters' brains, were both associated with dramatic improvements of survival, body weight gain, sensorimotor development and with decreased risk of epileptic seizures. Improvement of microglia activation status did not persist postnatally after doxycycline discontinuation; also, active brain infection remained unchanged by doxycycline. Altogether our data indicate that early microglia alteration, rather than brain CMV load per se, is instrumental in influencing survival and the neurological outcomes of CMV-infected rats, and suggest that microglia might participate in the neurological outcome of congenital CMV in humans. Furthermore this study represents a first proof-of-principle for the design of microglia-targeted preventive strategies in the context of congenital CMV infection of the brain.

12.
Methods Mol Biol ; 1677: 83-92, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28986866

RESUMEN

Transcription is the initial and generally the most sensitive step to cellular needs and environmental cues. Thus, it serves as a major mechanism controlling gene expression. Using reverse-transcription quantitative polymerase chain reaction technology (RT-qPCR), we will present how to quantify the transcriptional expression of NMDARs subunits during brain development and in both healthy and pathological conditions.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Animales , Encéfalo/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Humanos , ARN/genética , ARN/metabolismo , Receptores de N-Metil-D-Aspartato/genética
13.
J Vis Exp ; (130)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29286390

RESUMEN

Birth defects that involve the cerebral cortex - also known as malformations of cortical development (MCD) - are important causes of intellectual disability and account for 20-40% of drug-resistant epilepsy in childhood. High-resolution brain imaging has facilitated in vivo identification of a large group of MCD phenotypes. Despite the advances in brain imaging, genomic analysis and generation of animal models, a straightforward workflow to systematically prioritize candidate genes and to test functional effects of putative mutations is missing. To overcome this problem, an experimental strategy enabling the identification of novel causative genes for MCD was developed and validated. This strategy is based on identifying candidate genomic regions or genes via array-CGH or whole-exome sequencing and characterizing the effects of their inactivation or of overexpression of specific mutations in developing rodent brains via in utero electroporation. This approach led to the identification of the C6orf70 gene, encoding for a putative vesicular protein, to the pathogenesis of periventricular nodular heterotopia, a MCD caused by defective neuronal migration.


Asunto(s)
Encéfalo/patología , Hibridación Genómica Comparativa/métodos , Electroporación/métodos , Secuenciación del Exoma/métodos , Malformaciones del Desarrollo Cortical/genética , Animales , Química Encefálica , ADN/sangre , ADN/genética , ADN/aislamiento & purificación , Modelos Animales de Enfermedad , Femenino , Humanos , Malformaciones del Desarrollo Cortical/sangre , Malformaciones del Desarrollo Cortical/patología , Embarazo , Ratas
14.
Sci Rep ; 6: 33259, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27624926

RESUMEN

Polarity protein complexes function during polarized cell migration and a subset of these proteins localizes to the reoriented centrosome during this process. Despite these observations, the mechanisms behind the recruitment of these polarity complexes such as the aPKC/PAR6α complex to the centrosome are not well understood. Here we identify Hook2 as an interactor for the aPKC/PAR6α complex that functions to localize this complex at the centrosome. We first demonstrate that Hook2 is essential for the polarized Golgi re-orientation towards the migration front. Depletion of Hook2 results in a decrease of PAR6α at the centrosome during cell migration, while overexpression of Hook2 in cells induced the formation of aggresomes with the recruitment of PAR6α, aPKC and PAR3. In addition, we demonstrate that the interaction between the C-terminal domain of Hook2 and the aPKC-binding domain of PAR6α localizes PAR6α to the centrosome during cell migration. Our data suggests that Hook2, a microtubule binding protein, plays an important role in the regulation of PAR6α recruitment to the centrosome to bridge microtubules and the aPKC/PAR complex. This data reveals how some of the polarity protein complexes are recruited to the centrosome and might regulate pericentriolar and microtubule organization and potentially impact on polarized migration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Movimiento Celular/genética , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Proteína Quinasa C/genética , Animales , Polaridad Celular/genética , Centrosoma/metabolismo , Segregación Cromosómica/genética , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Humanos , Cuerpos de Inclusión/genética , Células MCF-7 , Microtúbulos/genética , Microtúbulos/metabolismo , Unión Proteica
15.
PLoS One ; 11(7): e0160176, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27472761

RESUMEN

BACKGROUND: Congenital cytomegalovirus infections are a leading cause of neurodevelopmental disorders in human and represent a major health care and socio-economical burden. In contrast with this medical importance, the pathophysiological events remain poorly known. Murine models of brain cytomegalovirus infection, mostly neonatal, have brought recent insights into the possible pathogenesis, with convergent evidence for the alteration and possible involvement of brain immune cells. OBJECTIVES AND METHODS: In order to confirm and expand those findings, particularly concerning the early developmental stages following infection of the fetal brain, we have created a model of in utero cytomegalovirus infection in the developing rat brain. Rat cytomegalovirus was injected intraventricularly at embryonic day 15 (E15) and the brains analyzed at various stages until the first postnatal day, using a combination of gene expression analysis, immunohistochemistry and multicolor flow cytometry experiments. RESULTS: Rat cytomegalovirus infection was increasingly seen in various brain areas including the choroid plexi and the ventricular and subventricular areas and was prominently detected in CD45low/int, CD11b+ microglial cells, in CD45high, CD11b+ cells of the myeloid lineage including macrophages, and in CD45+, CD11b- lymphocytes and non-B non-T cells. In parallel, rat cytomegalovirus infection of the developing rat brain rapidly triggered a cascade of pathophysiological events comprising: chemokines upregulation, including CCL2-4, 7 and 12; infiltration by peripheral cells including B-cells and monocytes at E17 and P1, and T-cells at P1; and microglia activation at E17 and P1. CONCLUSION: In line with previous findings in neonatal murine models and in human specimen, our study further suggests that neuroimmune alterations might play critical roles in the early stages following cytomegalovirus infection of the brain in utero. Further studies are now needed to determine which role, whether favorable or detrimental, those putative double-edge swords events actually play.


Asunto(s)
Encéfalo/embriología , Infecciones por Citomegalovirus/patología , Microglía/patología , Muromegalovirus/patogenicidad , Animales , Linaje de la Célula , Infecciones por Citomegalovirus/inmunología , Citometría de Flujo , Activación de Macrófagos , Microglía/inmunología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Eur J Hum Genet ; 24(6): 838-43, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26395558

RESUMEN

Semaphorins are a large family of secreted and membrane-associated proteins necessary for wiring of the brain. Semaphorin 5A (SEMA5A) acts as a bifunctional guidance cue, exerting both attractive and inhibitory effects on developing axons. Previous studies have suggested that SEMA5A could be a susceptibility gene for autism spectrum disorders (ASDs). We first identified a de novo translocation t(5;22)(p15.3;q11.21) in a patient with ASD and intellectual disability (ID). At the translocation breakpoint on chromosome 5, we observed a 861-kb deletion encompassing the end of the SEMA5A gene. We delineated the breakpoint by NGS and observed that no gene was disrupted on chromosome 22. We then used Sanger sequencing to search for deleterious variants affecting SEMA5A in 142 patients with ASD. We also identified two independent heterozygous variants located in a conserved functional domain of the protein. Both variants were maternally inherited and predicted as deleterious. Our genetic screens identified the first case of a de novo SEMA5A microdeletion in a patient with ASD and ID. Although our study alone cannot formally associate SEMA5A with susceptibility to ASD, it provides additional evidence that Semaphorin dysfunction could lead to ASD and ID. Further studies on Semaphorins are warranted to better understand the role of this family of genes in susceptibility to neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Deleción Cromosómica , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Trastorno del Espectro Autista/complicaciones , Trastorno del Espectro Autista/diagnóstico , Niño , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 22/genética , Cromosomas Humanos Par 5/genética , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/diagnóstico , Masculino , Herencia Paterna , Semaforinas , Translocación Genética
17.
Mol Biol Cell ; 22(23): 4549-62, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21998199

RESUMEN

Primary cilia originate from the centrosome and play essential roles in several cellular, developmental, and pathological processes, but the underlying mechanisms of ciliogenesis are not fully understood. Given the involvement of the adaptor protein Hook2 in centrosomal homeostasis and protein transport to pericentrosomal aggresomes, we explored its role in ciliogenesis. We found that in human retinal epithelial cells, Hook2 localizes at the Golgi apparatus and centrosome/basal body, a strategic partitioning for ciliogenesis. Of importance, Hook2 depletion disrupts ciliogenesis at a stage before the formation of the ciliary vesicle at the distal tip of the mother centriole. Using two hybrid and immunoprecipitation assays and a small interfering RNA strategy, we found that Hook2 interacts with and stabilizes pericentriolar material protein 1 (PCM1), which was reported to be essential for the recruitment of Rab8a, a GTPase that is believed to be crucial for membrane transport to the primary cilium. Of interest, GFP::Rab8a coimmunoprecipitates with endogenous Hook2 and PCM1. Finally, GFP::Rab8a can overcome Hook2 depletion, demonstrating a functional interaction between Hook2 and these two important regulators of ciliogenesis. The data indicate that Hook2 interacts with PCM1 in a complex that also contains Rab8a and regulates a limiting step required for further initiation of ciliogenesis after centriole maturation.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Epitelio Pigmentado de la Retina/citología , Secuencia de Aminoácidos , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Centrosoma/metabolismo , Cilios/metabolismo , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , ARN Interferente Pequeño , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA