Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Neurosci ; 45(3): 126-138, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36882009

RESUMEN

Alterations in the expression of genes encoding proteins involved in synapse formation, maturation, and function are a hallmark of many neurodevelopmental and psychiatric disorders. For example, there is reduced neocortical expression of the MET receptor tyrosine kinase (MET) transcript and protein in Autism Spectrum Disorder (ASD) and Rett syndrome. Preclinical in vivo and in vitro models manipulating MET signaling reveal that the receptor modulates excitatory synapse development and maturation in select forebrain circuits. The molecular adaptations underlying the altered synaptic development remain unknown. We performed a comparative mass spectrometry analysis of synaptosomes generated from the neocortex of wild type and Met null mice during the peak of synaptogenesis (postnatal day 14; data are available from ProteomeXchange with identifier PXD033204). The analyses revealed broad disruption of the developing synaptic proteome in the absence of MET, consistent with the localization of MET protein in pre- and postsynaptic compartments, including proteins associated with the neocortical synaptic MET interactome and those encoded by syndromic and ASD risk genes. In addition to an overrepresentation of altered proteins associated with the SNARE complex, multiple proteins in the ubiquitin-proteasome system and associated with the synaptic vesicle, as well as proteins that regulate actin filament organization and synaptic vesicle exocytosis/endocytosis, were disrupted. Taken together, the proteomic changes are consistent with structural and functional changes observed following alterations in MET signaling. We hypothesize that the molecular adaptations following Met deletion may reflect a general mechanism that produces circuit-specific molecular changes due to loss or reduction of synaptic signaling proteins.


Asunto(s)
Trastorno del Espectro Autista , Neocórtex , Ratones , Animales , Sinaptosomas/metabolismo , Proteoma/metabolismo , Trastorno del Espectro Autista/genética , Proteómica/métodos , Sinapsis/metabolismo
2.
Pediatr Res ; 94(2): 564-574, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36650307

RESUMEN

BACKGROUND: Although investigations have begun to differentiate biological and neurobiological responses to a variety of adversities, studies considering both endocrine and immune function in the same datasets are limited. METHODS: Associations between proximal (family functioning, caregiver depression, and anxiety) and distal (SES-D; socioeconomic disadvantage) early-life adversities with salivary inflammatory biomarkers (IL-1ß, IL-6, IL-8, and TNF-α) and hair HPA markers (cortisol, cortisone, and dehydroepiandrosterone) were examined in two samples of young U.S. children (N = 142; N = 145). RESULTS: Children exposed to higher SES-D had higher levels of TNF-α (B = 0.13, p = 0.011), IL-1ß (B = 0.10, p = 0.033), and DHEA (B = 0.16, p = 0.011). Higher family dysfunction was associated with higher cortisol (B = 0.08, p = 0.033) and cortisone (B = 0.05, p = 0.003). An interaction between SES-D and family dysfunction was observed for cortisol levels (p = 0.020) whereby children exposed to lower/average levels of SES-D exhibited a positive association between family dysfunction and cortisol levels, whereas children exposed to high levels of SES-D did not. These findings were partially replicated in the second sample. CONCLUSIONS: Our results indicate that these biological response systems may react differently to different forms of early-life adversity. IMPACT: Different forms of early-life adversity have varied stress signatures, and investigations of early-life adversities with inflammation and HPA markers are lacking. Children with higher socioeconomic disadvantage had higher TNF-α, IL-1ß, and DHEA. Higher family dysfunction was associated with higher hair cortisol and cortisone levels, and the association between family dysfunction and cortisol was moderated by socioeconomic disadvantage. Biological response systems (immune and endocrine) were differentially associated with distinct forms of early-life adversities.


Asunto(s)
Cortisona , Hidrocortisona , Humanos , Niño , Factor de Necrosis Tumoral alfa , Estrés Psicológico , Saliva , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Deshidroepiandrosterona
3.
Cereb Cortex ; 32(8): 1769-1786, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-34470051

RESUMEN

The molecular regulation of the temporal dynamics of circuit maturation is a key contributor to the emergence of normal structure-function relations. Developmental control of cortical MET receptor tyrosine kinase, expressed early postnatally in subpopulations of excitatory neurons, has a pronounced impact on the timing of glutamatergic synapse maturation and critical period plasticity. Here, we show that using a controllable overexpression (cto-Met) transgenic mouse, extending the duration of MET signaling after endogenous Met is switched off leads to altered molecular constitution of synaptic proteins, persistent activation of small GTPases Cdc42 and Rac1, and sustained inhibitory phosphorylation of cofilin. These molecular changes are accompanied by an increase in the density of immature dendritic spines, impaired cortical circuit maturation of prefrontal cortex layer 5 projection neurons, and altered laminar excitatory connectivity. Two photon in vivo imaging of dendritic spines reveals that cto-Met enhances de novo spine formation while inhibiting spine elimination. Extending MET signaling for two weeks in developing cortical circuits leads to pronounced repetitive activity and impaired social interactions in adult mice. Collectively, our data revealed that temporally controlled MET signaling as a critical mechanism for controlling cortical circuit development and emergence of normal behavior.


Asunto(s)
Neuronas , Sinapsis , Animales , Período Crítico Psicológico , Espinas Dendríticas/fisiología , Ratones , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Neuronas/fisiología , Sinapsis/fisiología
4.
J Neurosci ; 41(15): 3301-3306, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33597270

RESUMEN

Heterogeneity is defined as the quality or state of being diverse in character or content. This article summarizes the natural progression from my studies, reported in the first issue of the Journal of Neuroscience, that identified molecular heterogeneity in precursor cells of the developing primate cerebral cortex to the current state in which differences defined at the molecular, cellular, circuit, and systems levels are building data encyclopedias. The emphasis on heterogeneity has impacted many contributors in the field of developmental neuroscience, who have led a quest to determine the extent to which there is diversity, when it appears developmentally, and what heritable and nonheritable factors mediate nervous system assembly and function. Since the appearance of the article on progenitor cell heterogeneity in the inaugural issue of the Journal of Neuroscience, there have been continuous advances in technologies and data analytics that are contributing to a much better understanding of the origins of neurobiological and behavioral heterogeneity.


Asunto(s)
Ventrículos Cerebrales/citología , Células-Madre Neurales/fisiología , Neurogénesis , Neuroglía/fisiología , Animales , Ventrículos Cerebrales/crecimiento & desarrollo , Ventrículos Cerebrales/fisiología , Humanos , Células-Madre Neurales/citología , Neuroglía/citología
5.
Mol Psychiatry ; 26(8): 3723-3736, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-31900430

RESUMEN

Normal development of cortical circuits, including experience-dependent cortical maturation and plasticity, requires precise temporal regulation of gene expression and molecular signaling. Such regulation, and the concomitant impact on plasticity and critical periods, is hypothesized to be disrupted in neurodevelopmental disorders. A protein that may serve such a function is the MET receptor tyrosine kinase, which is tightly regulated developmentally in rodents and primates, and exhibits reduced cortical expression in autism spectrum disorder and Rett Syndrome. We found that the peak of MET expression in developing mouse cortex coincides with the heightened period of synaptogenesis, but is precipitously downregulated prior to extensive synapse pruning and certain peak periods of cortical plasticity. These results reflect a potential on-off regulatory synaptic mechanism for specific glutamatergic cortical circuits in which MET is enriched. In order to address the functional significance of the 'off' component of the proposed mechanism, we created a controllable transgenic mouse line that sustains cortical MET signaling. Continued MET expression in cortical excitatory neurons disrupted synaptic protein profiles, altered neuronal morphology, and impaired visual cortex circuit maturation and connectivity. Remarkably, sustained MET signaling eliminates monocular deprivation-induced ocular dominance plasticity during the normal cortical critical period; while ablating MET signaling leads to early closure of critical period plasticity. The results demonstrate a novel mechanism in which temporal regulation of a pleiotropic signaling protein underlies cortical circuit maturation and timing of cortical critical period, features that may be disrupted in neurodevelopmental disorders.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Plasticidad Neuronal , Proteínas Proto-Oncogénicas c-met , Animales , Trastorno del Espectro Autista , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-met/genética , Sinapsis
6.
Dev Psychobiol ; 64(7): e22332, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36282765

RESUMEN

Attentional biases to threat-related stimuli, such as fearful and angry facial expressions, are important to survival and emerge early in development. Infants demonstrate an attentional bias to fearful facial expressions by 5-7 months of age and an attentional bias toward anger by 3 years of age that are modulated by experiential factors. In a longitudinal study of 87 mother-infant dyads from families predominantly experiencing low income, we examined whether maternal stress and depressive symptoms were associated with trajectories of attentional biases to threat, assessed during an attention disengagement eye-tracking task when infants were 6-, 9-, and 12-month old. By 9 months, infants demonstrated a generalized bias toward threat (both fearful and angry facial expressions). Maternal perceived stress was associated with the trajectory of the bias toward angry facial expressions between 6 and 12 months. Specifically, infants of mothers with higher perceived stress exhibited a greater bias toward angry facial expressions at 6 months that decreased across the next 6 months, compared to infants of mothers with lower perceived stress who displayed an increased bias to angry facial expressions over this age range. Maternal depressive symptoms and stressful life events were not associated with trajectories of infant attentional bias to anger or fear. These findings highlight the role of maternal perceptions of stress in shaping developmental trajectories of threat-alerting systems.


Asunto(s)
Sesgo Atencional , Expresión Facial , Lactante , Femenino , Humanos , Estudios Longitudinales , Miedo , Ira
7.
J Pineal Res ; 70(2): e12705, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33210730

RESUMEN

Seasonal day length has been linked to the prevalence of mood disorders, and however, the mechanisms underlying this relationship remain unknown. Previous work in our laboratory has shown that developmental exposure to seasonal photoperiods has enduring effects on the activity of mouse dorsal raphe serotonergic neurons, their intrinsic electrical properties, as well as on depression and anxiety-related behaviors. Here we focus on the possible ionic mechanisms that underlie the observed programming of the electrophysiological properties of serotonin neurons, focusing on the twin-pore K + channels TREK-1 and TASK-1 that set resting membrane potential and regulate excitability. Pharmacological inhibition of TREK-1 significantly increased spike frequency in Short and Equinox photoperiods, but did not further elevate the firing rate in slices from Long photoperiod mice, suggesting that TREK-1 function is reduced in Long photoperiods. In contrast, inhibition of TASK-1 resulted in increases in firing rates across all photoperiods, suggesting that it contributes to setting excitability, but is not regulated by photoperiod. We then quantified Kcnk2 mRNA levels specifically in dorsal raphe 5-HT neurons using triple-label RNAscope. We found that Long photoperiod significantly reduced levels of Kcnk2 in serotonin neurons co-expressing Tph2, and Pet-1. Photoperiodic effects on the function and expression of TREK-1 were blocked in melatonin 1 receptor knockout (MT-1KO) mice, consistent with previous findings that MT-1 signaling is necessary for photoperiodic programming of dorsal raphe 5-HT neurons. Taken together these results indicate that photoperiodic regulation of TREK-1 expression and function plays a key role in photoperiodic programming the excitability of dorsal raphe 5-HT neurons.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Neuronas Serotoninérgicas/metabolismo , Animales , Electrofisiología , Femenino , Humanos , Masculino , Melatonina/metabolismo , Fotoperiodo , Canales de Potasio de Dominio Poro en Tándem/genética , Receptores de Melatonina/metabolismo , Serotonina/metabolismo
8.
Cereb Cortex ; 29(1): 189-201, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29190358

RESUMEN

The complex circuitry and cell-type diversity of the cerebral cortex are required for its high-level functions. The mechanisms underlying the diversification of cortical neurons during prenatal development have received substantial attention, but understanding of neuronal heterogeneity is more limited during later periods of cortical circuit maturation. To address this knowledge gap, connectivity analysis and molecular phenotyping of cortical neuron subtypes that express the developing synapse-enriched MET receptor tyrosine kinase were performed. Experiments used a MetGFP transgenic mouse line, combined with coexpression analysis of class-specific molecular markers and retrograde connectivity mapping. The results reveal that MET is expressed by a minor subset of subcerebral and a larger number of intratelencephalic projection neurons. Remarkably, MET is excluded from most layer 6 corticothalamic neurons. These findings are particularly relevant for understanding the maturation of discrete cortical circuits, given converging evidence that MET influences dendritic elaboration and glutamatergic synapse maturation. The data suggest that classically defined cortical projection classes can be further subdivided based on molecular characteristics that likely influence synaptic maturation and circuit wiring. Additionally, given that MET is classified as a high confidence autism risk gene, the data suggest that projection neuron subpopulations may be differentially vulnerable to disorder-associated genetic variation.


Asunto(s)
Red Nerviosa/metabolismo , Neuronas/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-met/biosíntesis , Corteza Somatosensorial/metabolismo , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Red Nerviosa/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-met/genética , Corteza Somatosensorial/crecimiento & desarrollo
10.
PLoS Genet ; 12(11): e1006425, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27846226

RESUMEN

Sexual dimorphism in common disease is pervasive, including a dramatic male preponderance in autism spectrum disorders (ASDs). Potential genetic explanations include a liability threshold model requiring increased polymorphism risk in females, sex-limited X-chromosome contribution, gene-environment interaction driven by differences in hormonal milieu, risk influenced by genes sex-differentially expressed in early brain development, or contribution from general mechanisms of sexual dimorphism shared with secondary sex characteristics. Utilizing a large single nucleotide polymorphism (SNP) dataset, we identify distinct sex-specific genome-wide significant loci. We investigate genetic hypotheses and find no evidence for increased genetic risk load in females, but evidence for sex heterogeneity on the X chromosome, and contribution of sex-heterogeneous SNPs for anthropometric traits to ASD risk. Thus, our results support pleiotropy between secondary sex characteristic determination and ASDs, providing a biological basis for sex differences in ASDs and implicating non brain-limited mechanisms.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Cromosomas Humanos X/genética , Trastorno del Espectro Autista/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Trastornos Generalizados del Desarrollo Infantil/patología , Femenino , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética , Caracteres Sexuales
11.
J Neurosci ; 37(45): 10783-10791, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118206

RESUMEN

Adversity in early childhood exerts an enduring impact on mental and physical health, academic achievement, lifetime productivity, and the probability of interfacing with the criminal justice system. More science is needed to understand how the brain is affected by early life stress (ELS), which produces excessive activation of stress response systems broadly throughout the child's body (toxic stress). Our research examines the importance of sex, timing and type of stress exposure, and critical periods for intervention in various brain systems across species. Neglect (the absence of sensitive and responsive caregiving) or disrupted interaction with offspring induces robust, lasting consequences in mice, monkeys, and humans. Complementary assessment of internalizing disorders and brain imaging in children suggests that early adversity can interfere with white matter development in key brain regions, which may increase risk for emotional difficulties in the long term. Neural circuits that are most plastic during ELS exposure in monkeys sustain the greatest change in gene expression, offering a mechanism whereby stress timing might lead to markedly different long-term behaviors. Rodent models reveal that disrupted maternal-infant interactions yield metabolic and behavioral outcomes often differing by sex. Moreover, ELS may further accelerate or delay critical periods of development, which reflect GABA circuit maturation, BDNF, and circadian Clock genes. Such factors are associated with several mental disorders and may contribute to a premature closure of plastic windows for intervention following ELS. Together, complementary cross-species studies are elucidating principles of adaptation to adversity in early childhood with molecular, cellular, and whole organism resolution.


Asunto(s)
Maltrato a los Niños/psicología , Discapacidades del Desarrollo/etiología , Medio Social , Adulto , Niño , Preescolar , Discapacidades del Desarrollo/psicología , Cuidados en el Hogar de Adopción/psicología , Humanos , Estrés Psicológico/psicología
12.
J Neurosci ; 36(23): 6258-68, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27277803

RESUMEN

UNLABELLED: Trauma- and stress-related disorders are clinically heterogeneous and associated with substantial genetic risk. Understanding the biological origins of heterogeneity of key intermediate phenotypes such as cognition and emotion can provide novel mechanistic insights into disorder pathogenesis. Performing quantitative genetics in animal models is a tractable strategy for examining both the genetic basis of intermediate phenotypes and functional testing of candidate quantitative traits genes (QTGs). Here, existing and newly collected data were used for collaborative genome-wide mapping of cued fear acquisition and expression in 65 mouse strains from the BXD genetic reference panel. For fear acquisition, we identified a significant locus on chromosome (Chr) 10 and eight suggestive loci on Chr 2, 4, 5, 11, 13, and 15. For fear expression, we identified one significant and another highly suggestive locus on Chr 13, as well as four suggestive loci on Chr 10, 11, and X. Across these loci, 60 putative QTGs were identified. The quantitative trait locus on distal Chr 13 contained a single, highly promising gene at the location of the peak likelihood ratio statistic score. The gene, hyperpolarization-activated cyclic nucleotide-gated channel 1 (Hcn1), regulates neuronal excitability. Validation experiments using behavioral pharmacology revealed that functional Hcn channels in the basolateral amygdala are necessary for conditioned fear acquisition and expression. Hcn1, together with the other candidate QTGs, thus provide new targets for neurobiological and treatment studies of fear learning and trauma- and stress-related disorders. SIGNIFICANCE STATEMENT: There is a knowledge gap in understanding the genetic contributions to behavioral heterogeneity in typical and atypical populations. Mouse genetic reference panels (GRPs) provide one approach for identifying genetic sources of variation. Here, we identified three loci for conditioned fear acquisition and expression in a mouse GRP. Each locus contained candidate quantitative trait genes (QTGs). One locus had a single QTG, Hcn1 (hyperpolarization-activated cyclic nucleotide-gated channel 1), which has been implicated in neuronal excitability and learning. This discovery was validated using behavioral pharmacology, revealing that Hcn channels in the basolateral amygdala are required for fear acquisition and expression. The study thus identifies novel candidate QTGs that may contribute to variation in emotional learning and highlight the utility of mouse GRPs for the identification of genes underlying complex traits.


Asunto(s)
Mapeo Cromosómico , Condicionamiento Clásico/fisiología , Miedo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales de Potasio/genética , Sitios de Carácter Cuantitativo/genética , Análisis de Varianza , Animales , Fármacos Cardiovasculares/farmacología , Corteza Cerebral/metabolismo , Cromosomas Humanos Par 13/genética , Reacción Cataléptica de Congelación/fisiología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos , Fenotipo , Pirimidinas/farmacología
13.
J Neurosci ; 36(31): 8200-9, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27488639

RESUMEN

UNLABELLED: The stria vascularis is a nonsensory structure that is essential for auditory hair cell function by maintaining potassium concentration of the scala media. During mouse embryonic development, a subpopulation of neural crest cell-derived melanocytes migrates and incorporates into a subregion of the cochlear epithelium, forming the intermediate cell layer of the stria vascularis. The relation of this developmental process to stria vascularis function is currently unknown. In characterizing the molecular differentiation of developing peripheral auditory structures, we discovered that hepatocyte growth factor (Hgf) is expressed in the future stria vascularis of the cochlear epithelium. Its receptor tyrosine kinase, c-Met, is expressed in the cochlear epithelium and melanocyte-derived intermediate cells in the stria vascularis. Genetic dissection of HGF signaling via c-MET reveals that the incorporation of the melanocytes into the future stria vascularis of the cochlear duct requires c-MET signaling. In addition, inactivation of either the ligand or receptor developmentally resulted in a profound hearing loss at young adult stages. These results suggest a novel connection between HGF signaling and deafness via melanocyte deficiencies. SIGNIFICANCE STATEMENT: We found the roles of hepatocyte growth factor (HGF) signaling in stria vascularis development for the first time and that lack of HGF signaling in the inner ear leads to profound hearing loss in the mouse. Our findings reveal a novel mechanism that may underlie human deafness DFNB39 and DFNB97. Our findings reveal an additional example of context-dependent c-MET signaling diversity, required here for proper cellular invasion developmentally that is essential for specific aspects of auditory-related organogenesis.


Asunto(s)
Pérdida Auditiva/metabolismo , Audición , Factor de Crecimiento de Hepatocito/metabolismo , Melanocitos/metabolismo , Melanocitos/fisiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Cóclea/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Pérdida Auditiva/embriología , Pérdida Auditiva/patología , Masculino , Ratones , Transducción de Señal , Estría Vascular/patología
14.
Nature ; 472(7343): 347-50, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21512572

RESUMEN

Serotonin (5-hydroxytryptamine or 5-HT) is thought to regulate neurodevelopmental processes through maternal-fetal interactions that have long-term mental health implications. It is thought that beyond fetal 5-HT neurons there are significant maternal contributions to fetal 5-HT during pregnancy but this has not been tested empirically. To examine putative central and peripheral sources of embryonic brain 5-HT, we used Pet1(-/-) (also called Fev) mice in which most dorsal raphe neurons lack 5-HT. We detected previously unknown differences in accumulation of 5-HT between the forebrain and hindbrain during early and late fetal stages, through an exogenous source of 5-HT which is not of maternal origin. Using additional genetic strategies, a new technology for studying placental biology ex vivo and direct manipulation of placental neosynthesis, we investigated the nature of this exogenous source. We uncovered a placental 5-HT synthetic pathway from a maternal tryptophan precursor in both mice and humans. This study reveals a new, direct role for placental metabolic pathways in modulating fetal brain development and indicates that maternal-placental-fetal interactions could underlie the pronounced impact of 5-HT on long-lasting mental health outcomes.


Asunto(s)
Feto/metabolismo , Intercambio Materno-Fetal/fisiología , Placenta/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Serotonina/biosíntesis , Animales , Embrión de Mamíferos/metabolismo , Femenino , Feto/embriología , Humanos , Técnicas In Vitro , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal , Núcleos del Rafe/citología , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Serotonina/análisis , Serotonina/metabolismo , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
15.
J Neurosci ; 34(49): 16166-79, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471559

RESUMEN

The MET receptor tyrosine kinase (RTK), implicated in risk for autism spectrum disorder (ASD) and in functional and structural circuit integrity in humans, is a temporally and spatially regulated receptor enriched in dorsal pallial-derived structures during mouse forebrain development. Here we report that loss or gain of function of MET in vitro or in vivo leads to changes, opposite in nature, in dendritic complexity, spine morphogenesis, and the timing of glutamatergic synapse maturation onto hippocampus CA1 neurons. Consistent with the morphological and biochemical changes, deletion of Met in mutant mice results in precocious maturation of excitatory synapse, as indicated by a reduction of the proportion of silent synapses, a faster GluN2A subunit switch, and an enhanced acquisition of AMPA receptors at synaptic sites. Thus, MET-mediated signaling appears to serve as a mechanism for controlling the timing of neuronal growth and functional maturation. These studies suggest that mistimed maturation of glutamatergic synapses leads to the aberrant neural circuits that may be associated with ASD risk.


Asunto(s)
Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Dendritas/metabolismo , Espinas Dendríticas/fisiología , Neurogénesis/fisiología , Proteínas Proto-Oncogénicas c-met/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/enzimología , Región CA1 Hipocampal/crecimiento & desarrollo , Ratones , Mutación , Neuronas/citología , Neuronas/fisiología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-met/genética , Receptores AMPA/metabolismo , Transducción de Señal/fisiología
16.
Nature ; 461(7263): 524-8, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19759535

RESUMEN

The cerebral cortex is a laminated sheet of neurons composed of the arrays of intersecting radial columns. During development, excitatory projection neurons originating from the proliferative units at the ventricular surface of the embryonic cerebral vesicles migrate along elongated radial glial fibres to form a cellular infrastructure of radial (vertical) ontogenetic columns in the overlaying cortical plate. However, a subpopulation of these clonally related neurons also undergoes a short lateral shift and transfers from their parental to the neighbouring radial glial fibres, and intermixes with neurons originating from neighbouring proliferative units. This columnar organization acts as the primary information processing unit in the cortex. The molecular mechanisms, role and significance of this lateral dispersion for cortical development are not understood. Here we show that an Eph receptor A (EphA) and ephrin A (Efna) signalling-dependent shift in the allocation of clonally related neurons is essential for the proper assembly of cortical columns. In contrast to the relatively uniform labelling of the developing cortical plate by various molecular markers and retrograde tracers in wild-type mice, we found alternating labelling of columnar compartments in Efna knockout mice that are caused by impaired lateral dispersion of migrating neurons rather than by altered cell production or death. Furthermore, in utero electroporation showed that lateral dispersion depends on the expression levels of EphAs and ephrin-As during neuronal migration. This so far unrecognized mechanism for lateral neuronal dispersion seems to be essential for the proper intermixing of neuronal types in the cortical columns, which, when disrupted, might contribute to neuropsychiatric disorders associated with abnormal columnar organization.


Asunto(s)
Movimiento Celular , Corteza Cerebral/embriología , Efrinas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal , Animales , Corteza Cerebral/anatomía & histología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Efrinas/deficiencia , Efrinas/genética , Ratones , Ratones Noqueados , Neocórtex/citología , Neocórtex/metabolismo , Organogénesis , Ratas , Receptores de la Familia Eph/deficiencia , Receptores de la Familia Eph/genética
17.
Nat Rev Neurosci ; 10(4): 303-12, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19277053

RESUMEN

The effects of prenatal exposure to drugs on brain development are complex and are modulated by the timing, dose and route of drug exposure. It is difficult to assess these effects in clinical cohorts as these are beset with problems such as multiple exposures and difficulties in documenting use patterns. This can lead to misinterpretation of research findings by the general public, the media and policy makers, who may mistakenly assume that the legal status of a drug correlates with its biological impact on fetal brain development and long-term clinical outcomes. It is important to close the gap between what science tells us about the impact of prenatal drug exposure on the fetus and the mother and what we do programmatically with regard to at-risk populations.


Asunto(s)
Encéfalo/efectos de los fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Complicaciones del Embarazo/inducido químicamente , Efectos Tardíos de la Exposición Prenatal , Alcoholes/toxicidad , Anfetamina/toxicidad , Antidepresivos/toxicidad , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Niño , Cocaína Crack/toxicidad , Femenino , Humanos , Intercambio Materno-Fetal , Metanfetamina/toxicidad , Nicotina/toxicidad , Embarazo , Trastornos Relacionados con Sustancias
18.
Cereb Cortex ; 23(4): 775-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22490549

RESUMEN

Auditory stimulus representations are dynamically maintained by ascending and descending projections linking the auditory cortex (Actx), medial geniculate body (MGB), and inferior colliculus. Although the extent and topographic specificity of descending auditory corticofugal projections can equal or surpass that of ascending corticopetal projections, little is known about the molecular mechanisms that guide their development. Here, we used in utero gene electroporation to examine the role of EphA receptor signaling in the development of corticothalamic (CT) and corticocollicular connections. Early in postnatal development, CT axons were restricted to a deep dorsal zone (DDZ) within the MGB that expressed low levels of the ephrin-A ligand. By hearing onset, CT axons had innervated surrounding regions of MGB in control-electroporated mice but remained fixed within the DDZ in mice overexpressing EphA7. In vivo neurophysiological recordings demonstrated a corresponding reduction in spontaneous firing rate, but no changes in sound-evoked responsiveness within MGB regions deprived of CT innervation. Structural and functional CT disruption occurred without gross alterations in thalamocortical connectivity. These data demonstrate a potential role for EphA/ephrin-A signaling in the initial guidance of corticofugal axons and suggest that "genetic rewiring" may represent a useful functional tool to alter cortical feedback without silencing Actx.


Asunto(s)
Corteza Auditiva , Vías Auditivas/fisiología , Mapeo Encefálico , Cuerpos Geniculados/fisiología , Receptor EphA7/metabolismo , Transducción de Señal/fisiología , Estimulación Acústica , Factores de Edad , Aminoácidos , Animales , Animales Recién Nacidos , Corteza Auditiva/embriología , Corteza Auditiva/crecimiento & desarrollo , Corteza Auditiva/metabolismo , Axones/fisiología , Electroencefalografía , Electroporación , Embrión de Mamíferos , Potenciales Evocados Auditivos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo , Receptor EphA7/genética , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
19.
Genes Brain Behav ; 23(2): e12892, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38560770

RESUMEN

Mutations in CHD8 are one of the highest genetic risk factors for autism spectrum disorder. Studies in mice that investigate underlying mechanisms have shown Chd8 haploinsufficient mice display some trait disruptions that mimic clinical phenotypes, although inconsistencies have been reported in some traits across different models on the same strain background. One source of variation across studies may be the impact of Chd8 haploinsufficiency on maternal-offspring interactions. While differences in maternal care as a function of Chd8 genotype have not been studied directly, a previous study showed that pup survival was reduced when reared by Chd8 heterozygous dams compared with wild-type (WT) dams, suggesting altered maternal care as a function of Chd8 genotype. Through systematic observation of the C57BL/6 strain, we first determined the impact of Chd8 haploinsufficiency in the offspring on WT maternal care frequencies across preweaning development. We next determined the impact of maternal Chd8 haploinsufficiency on pup care. Compared with litters with all WT offspring, WT dams exhibited less frequent maternal behaviors toward litters consisting of offspring with mixed Chd8 genotypes, particularly during postnatal week 1. Dam Chd8 haploinsufficiency decreased litter survival and increased active maternal care also during postnatal week 1. Determining the impact of Chd8 haploinsufficiency on early life experiences provides an important foundation for interpreting offspring outcomes and determining mechanisms that underlie heterogeneous phenotypes.


Asunto(s)
Trastorno del Espectro Autista , Animales , Femenino , Ratones , Trastorno del Espectro Autista/genética , Genotipo , Haploinsuficiencia , Ratones Endogámicos C57BL , Fenotipo
20.
bioRxiv ; 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36778231

RESUMEN

Cognitive impairment is a common phenotype of neurodevelopmental disorders, but how these deficits arise remains elusive. Determining the onset of discrete cognitive capabilities facilitates studies in probing mechanisms underlying their emergence. The present study analyzed the emergence of contextual fear memory persistence (7-day memory retention) and remote memory (30-day memory retention). There was a rapid transition from postnatal day (P) 20 to P21, in which memory persistence emerged in C57Bl/6J male and female mice. Remote memory was present at P23, but expression was not robust compared to pubertal and adult mice. Previous studies reported that following deletion of the MET receptor tyrosine kinase (MET), there are fear memory deficits in adult mice and the timing of critical period plasticity is altered in the developing visual cortex, positioning MET as a regulator for onset of contextual fear memory. Sustaining Met past the normal window of peak cortical expression or deleting Met, however, did not alter the timing of emergence of persistence or remote memory capabilities during development. Fear memory in young adults, however, was disrupted. Remarkably, compared to homecage controls, the number of FOS-expressing infragranular neurons in medial prefrontal cortex (mPFC) did not increase from contextual memory formation recall of fear conditioning at P35 but exhibited enhanced activation at P90 in male and female mice. Additionally, MET-expressing neurons were preferentially recruited at P90 compared to P35 during fear memory expression. The studies demonstrate a developmental profile of contextual fear memory capabilities. Further, developmental disruption of Met leads to a delayed functional deficit that arises in young adulthood, correlated with an increase of mPFC neuron activation during fear memory recall.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA