Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neurobiol Dis ; 146: 105118, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33031903

RESUMO

Fragile X syndrome (FXS), a neurodevelopmental disorder with autistic features, is caused by the loss of the fragile X mental retardation protein. Sex-specific differences in the clinical profile have been observed in FXS patients, but few studies have directly compared males and females in rodent models of FXS. To address this, we performed electroencephalography (EEG) recordings and a battery of autism-related behavioral tasks on juvenile and young adult Fmr1 knockout (KO) rats. EEG analysis demonstrated that compared to wild-type, male Fmr1 KO rats showed an increase in gamma frequency band power in the frontal cortex during the sleep-like immobile state, and both male and female KO rats failed to show an increase in delta frequency power in the sleep-like state, as observed in wild-type rats. Previous studies of EEG profiles in FXS subjects also reported abnormally increased gamma frequency band power, highlighting this parameter as a potential translatable biomarker. Both male and female Fmr1 KO rats displayed reduced exploratory behaviors in the center zone of the open field test, and increased distance travelled in an analysis of 24-h home cage activity, an effect that was more prominent during the nocturnal phase. Reduced wins against wild-type opponents in the tube test of social dominance was seen in both sexes. In contrast, increased repetitive behaviors in the wood chew test was observed in male but not female KO rats, while increased freezing in a fear conditioning test was observed only in the female KO rats. Our findings highlight sex differences between male and female Fmr1 KO rats, and indicate that the rat model of FXS could be a useful tool for the development of new therapeutics for treating this debilitating neurodevelopmental disorder.


Assuntos
Córtex Auditivo/fisiopatologia , Transtorno Autístico/fisiopatologia , Comportamento Animal/fisiologia , Síndrome do Cromossomo X Frágil/fisiopatologia , Estimulação Acústica/métodos , Animais , Ansiedade/fisiopatologia , Córtex Auditivo/metabolismo , Transtorno do Espectro Autista/metabolismo , Transtorno Autístico/metabolismo , Modelos Animais de Doenças , Eletroencefalografia/métodos , Comportamento Exploratório/fisiologia , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Ratos
2.
Hum Mol Genet ; 24(21): 5977-84, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26243794

RESUMO

Recent advances in techniques for manipulating genomes have allowed the generation of transgenic animals other than mice. These new models enable cross-mammalian comparison of neurological disease from core cellular pathophysiology to circuit and behavioural endophenotypes. Moreover they will enable us to directly test whether common cellular dysfunction or behavioural outcomes of a genetic mutation are more conserved across species. Using a new rat model of Fragile X Syndrome, we report that Fmr1 knockout (KO) rats exhibit elevated basal protein synthesis and an increase in mGluR-dependent long-term depression in CA1 of the hippocampus that is independent of new protein synthesis. These defects in plasticity are accompanied by an increase in dendritic spine density selectively in apical dendrites and subtle changes in dendritic spine morphology of CA1 pyramidal neurons. Behaviourally, Fmr1 KO rats show deficits in hippocampal-dependent, but not hippocampal-independent, forms of associative recognition memory indicating that the loss of fragile X mental retardation protein (FMRP) causes defects in episodic-like memory. In contrast to previous reports from mice, Fmr1 KO rats show no deficits in spatial reference memory reversal learning. One-trial spatial learning in a delayed matching to place water maze task was also not affected by the loss of FMRP in rats. This is the first evidence for conservation across mammalian species of cellular and physiological hippocampal phenotypes associated with the loss of FMRP. Furthermore, while key cellular phenotypes are conserved they manifest in distinct behavioural dysfunction. Finally, our data reveal novel information about the selective role of FMRP in hippocampus-dependent associative memory.


Assuntos
Modelos Animais de Doenças , Síndrome do Cromossomo X Frágil/fisiopatologia , Hipocampo/fisiopatologia , Animais , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Técnicas de Inativação de Genes , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/genética , Transtornos da Memória/fisiopatologia , Plasticidade Neuronal , Células Piramidais/patologia , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
3.
Hum Mol Genet ; 21(10): 2143-56, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22328088

RESUMO

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and results from the loss of the fragile X mental retardation protein (FMRP). Many fragile X-related cognitive and behavioral features emerge during childhood and are associated with abnormal synaptic and cellular organization of the cerebral cortex. Identifying the roles of FMRP in cortical development will provide a basis for understanding the pathogenesis of the syndrome. However, how the loss of FMRP influences the developmental trajectory of cortical maturation remains unclear. We took advantage of the stereotyped and well-characterized development of the murine primary somatosensory cortex to examine cortical maturation during a time-window that corresponds to late embryonic and early postnatal development in the human. In the Fmr1 knockout mouse, we find a delay in somatosensory map formation, alterations in the morphology profile of dendrites and spines of layer 4 neurons and a decrease in the synaptic levels of proteins involved in glutamate receptor signaling at times corresponding to the highest levels of FMRP expression. In contrast, cortical arealization, synaptic density in layer 4 and early postnatal regulation of mRNAs encoding synaptic proteins are not altered in Fmr1 knockout mice. The specificity of the developmental delay in Fmr1 knockout mice indicates that the loss of FMRP does not result in a general stalling of cerebral cortex maturation. Instead, our results suggest that inaccurate timing of developmental processes caused by the loss of FMRP may lead to alterations in neural circuitry that underlie behavioral and cognitive dysfunctions associated with FXS.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/metabolismo , Córtex Somatossensorial/metabolismo , Animais , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica , RNA Mensageiro/metabolismo
4.
Front Syst Neurosci ; 17: 1229627, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38075533

RESUMO

Cerebellar networks have traditionally been linked to sensorimotor control. However, a large body of evidence suggests that cerebellar functions extend to non-motor realms, such as fear-based emotional processing and that these functions are supported by interactions with a wide range of brain structures. Research related to the cerebellar contributions to emotional processing has focussed primarily on the use of well-constrained conditioning paradigms in both human and non-human subjects. From these studies, cerebellar circuits appear to be critically involved in both conditioned and unconditioned responses to threatening stimuli in addition to encoding and storage of fear memory. It has been hypothesised that the computational mechanism underlying this contribution may involve internal models, where errors between actual and expected outcomes are computed within the circuitry of the cerebellum. From a clinical perspective, cerebellar abnormalities have been consistently linked to neurodevelopmental disorders, including autism. Importantly, atypical adaptive behaviour and heightened anxiety are also common amongst autistic individuals. In this review, we provide an overview of the current anatomical, physiological and theoretical understanding of cerebellar contributions to fear-based emotional processing to foster further insights into the neural circuitry underlying emotional dysregulation observed in people with autism.

5.
Learn Mem ; 18(1): 39-48, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21177378

RESUMO

Loss of the Fragile X mental retardation protein (FMRP) is associated with presumed postsynaptic deficits in mouse models of Fragile X syndrome. However, the possible presynaptic roles of FMRP in learning-related plasticity have received little attention. As a result, the mechanisms whereby FMRP influences synaptic function remain poorly understood. To investigate the cellular locus of the effects of FMRP on synaptic plasticity, we cloned the Aplysia homolog of FMRP and find it to be highly expressed in neurons. By selectively down-regulating FMRP in individual Aplysia neurons at the sensory-to-motor neuron synapse reconstituted in co-cultures, we demonstrate that FMRP functions both pre- and postsynaptically to constrain the expression of long-term synaptic depression induced by repeated pulses of FMRF-amide. In contrast, FMRP has little to no effect on long-term synaptic facilitation induced by repeated pulses of serotonin. Since other components of signaling pathways involved in plasticity appear to be conserved between Aplysia and mammalian neurons, our findings suggest that FMRP can participate in both pre- and postsynaptic regulation of enduring synaptic plasticity that underlies the storage of certain types of long-term memory.


Assuntos
Aplysia/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Potenciação de Longa Duração/fisiologia , Neurônios Motores/fisiologia , Terminações Pré-Sinápticas/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Aplysia/efeitos dos fármacos , Aplysia/genética , Aplysia/metabolismo , Células Cultivadas , Clonagem Molecular/métodos , Técnicas de Cocultura , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , FMRFamida/farmacologia , Proteína do X Frágil da Deficiência Intelectual/genética , Potenciação de Longa Duração/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Microscopia Confocal , Neurônios Motores/efeitos dos fármacos , Mutação/genética , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp/métodos , Terminações Pré-Sinápticas/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Serotonina/farmacologia
6.
Trends Neurosci ; 45(3): 171-172, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34998618

RESUMO

Advances in genetic technologies have facilitated the development of new animal models of neurodevelopmental disorders (NDDs), enabling cross-species validation of disease-related phenotypes and exploration of species-specific behaviours. In a recent study, Berg et al. used a rat model of Angelman Syndrome (AS) to identify Ube3a-dependent social behaviours, highlighting potential cross-species convergence and divergence between rodent models.


Assuntos
Síndrome de Angelman , Transtornos do Neurodesenvolvimento , Síndrome de Angelman/genética , Animais , Modelos Animais de Doenças , Humanos , Fenótipo , Ratos , Ubiquitina-Proteína Ligases/genética
7.
Brain Commun ; 4(6): fcac263, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36349120

RESUMO

Mutations in the SYNGAP1 gene are one of the common predictors of neurodevelopmental disorders, commonly resulting in individuals developing autism, intellectual disability, epilepsy, and sleep deficits. EEG recordings in neurodevelopmental disorders show potential to identify clinically translatable biomarkers to both diagnose and track the progress of novel therapeutic strategies, as well as providing insight into underlying pathological mechanisms. In a rat model of SYNGAP1 haploinsufficiency in which the exons encoding the calcium/lipid binding and GTPase-activating protein domains have been deleted (Syngap+/Δ-GAP ), we analysed the duration and occurrence of wake, non-rapid eye movement and rapid eye movement brain states during 6 h multi-electrode EEG recordings. We find that although Syngap+/Δ-GAP animals spend an equivalent percent time in wake and sleep states, they have an abnormal brain state distribution as the number of wake and non-rapid eye movement bouts are reduced and there is an increase in the average duration of both wake and non-rapid eye movement epochs. We perform connectivity analysis by calculating the average imaginary coherence between electrode pairs at varying distance thresholds during these states. In group averages from pairs of electrodes at short distances from each other, a clear reduction in connectivity during non-rapid eye movement is present between 11.5 Hz and 29.5 Hz, a frequency range that overlaps with sleep spindles, oscillatory phenomena thought to be important for normal brain function and memory consolidation. Sleep abnormalities were mostly uncorrelated to the electrophysiological signature of absence seizures, spike and wave discharges, as was the imaginary coherence deficit. Sleep spindles occurrence, amplitude, power and spread across multiple electrodes were not reduced in Syngap+/Δ-GAP rats, with only a small decrease in duration detected. Nonetheless, by analysing the dynamic imaginary coherence during sleep spindles, we found a reduction in high-connectivity instances between short-distance electrode pairs. Finally comparing the dynamic imaginary coherence during sleep spindles between individual electrode pairs, we identified a group of channels over the right somatosensory, association and visual cortices that have a significant reduction in connectivity during sleep spindles in mutant animals. This matched a significant reduction in connectivity during spindles when averaged regional comparisons were made. These data suggest that Syngap+/Δ-GAP rats have altered brain state dynamics and EEG connectivity, which may have clinical relevance for SYNGAP1 haploinsufficiency in humans.

8.
Biochem Soc Trans ; 38(2): 507-10, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20298211

RESUMO

FXS (Fragile X syndrome) is the most common genetically inherited form of cognitive impairment. The predominant cause of the syndrome is the loss of a single protein, FMRP (Fragile X mental retardation protein). Many of the cognitive and behavioural features found in Fragile X individuals emerge during childhood and are associated with abnormal organization of cortical connections. However, although FMRP is expressed as early as embryogenesis, relatively little is known about its roles during development or how this may influence FXS phenotypes in adulthood. The present review focuses specifically on the evidence for the functions of FMRP during embryonic and early postnatal development. The current knowledge of the role of FMRP in FXS will be briefly summarized before addressing how alterations in the formation and refinement of neuronal connections and synaptic function that result from the loss of FMRP may in turn influence behaviours that are expressed during the first few postnatal weeks. I will then briefly highlight some outstanding questions about the developmental roles of FMRP and their possible relationship to symptoms found in adults with FXS.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/fisiologia , Crescimento e Desenvolvimento/genética , Adulto , Fatores Etários , Animais , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Humanos , Rede Nervosa/embriologia , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Fenótipo
9.
Elife ; 92020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31939740

RESUMO

SynGAP is a postsynaptic density (PSD) protein that binds to PDZ domains of the scaffold protein PSD-95. We previously reported that heterozygous deletion of Syngap1 in mice is correlated with increased steady-state levels of other key PSD proteins that bind PSD-95, although the level of PSD-95 remains constant (Walkup et al., 2016). For example, the ratio to PSD-95 of Transmembrane AMPA-Receptor-associated Proteins (TARPs), which mediate binding of AMPA-type glutamate receptors to PSD-95, was increased in young Syngap1+/-mice. Here we show that only females and not males show a highly significant correlation between an increase in TARP and a decrease in synGAP in the PSDs of Syngap1+/-rodents. The data reveal a sex difference in the adaptation of the PSD scaffold to synGAP haploinsufficiency.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Haploinsuficiência , Densidade Pós-Sináptica/metabolismo , Animais , Sistemas CRISPR-Cas , Feminino , Proteínas Ativadoras de GTPase/genética , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Fatores Sexuais
10.
J Neurosci ; 28(49): 13028-37, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19052194

RESUMO

We have previously reported that mGluR5 signaling via PLC-beta1 regulates the development of whisker patterns within S1 (barrel) cortex of mice (Hannan et al., 2001). However, whether these defects arise from the loss of postsynaptic mGluR5 signaling, and whether the level of mGluR5 is important for barrel formation, was not examined. Furthermore, whether mGluR5 regulates other developmental processes that occur before or after barrel development is not known. We now show that mGluR5 is present postsynaptically at thalamocortical synapses during barrel formation. In addition, Mglur5(+/-) mice exhibit normal TCA patch formation but reduced cellular segregation in layer 4, indicating a dose-dependent role for mGluR5 in the regulation of pattern formation. Furthermore Mglur5(-/-) and Mglur5(+/-) mice display normal cortical arealization, layer formation, and size of PMBSF indicating the defects within S1 do not result from general abnormalities of cortical mapping during earlier stages of development. At P21 layer 4 neurons from Mglur5(-/-) and Mglur5(+/-) mice show a significant reduction in spine density but normal dendritic complexity compared with Mglur5(+/+) mice indicating a role in synaptogenesis during cortical development. Finally, mGluR5 regulates pattern formation throughout the trigeminal system of mice as the representation of the AS whiskers in the PrV, VpM, and S1 cortex was disrupted in Mglur5(-/-) mice. Together these data indicate a key role for mGluR5 at both early and late stages of neuronal development in the trigeminal system of mice.


Assuntos
Ácido Glutâmico/metabolismo , Neurogênese/genética , Receptores de Glutamato Metabotrópico/genética , Córtex Somatossensorial/anormalidades , Córtex Somatossensorial/crescimento & desenvolvimento , Sinapses/metabolismo , Vias Aferentes/anormalidades , Vias Aferentes/crescimento & desenvolvimento , Vias Aferentes/metabolismo , Animais , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Vias Neurais/anormalidades , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Receptor de Glutamato Metabotrópico 5 , Córtex Somatossensorial/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica/genética , Nervo Trigêmeo/anormalidades , Nervo Trigêmeo/crescimento & desenvolvimento , Nervo Trigêmeo/metabolismo , Núcleos Ventrais do Tálamo/anormalidades , Núcleos Ventrais do Tálamo/crescimento & desenvolvimento , Núcleos Ventrais do Tálamo/metabolismo , Vibrissas/inervação
11.
Sci Transl Med ; 11(494)2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31142675

RESUMO

Fragile X Syndrome (FXS) is one of the most common monogenic forms of autism and intellectual disability. Preclinical studies in animal models have highlighted the potential of pharmaceutical intervention strategies for alleviating the symptoms of FXS. However, whether treatment strategies can be tailored to developmental time windows that define the emergence of particular phenotypes is unknown. Similarly, whether a brief, early intervention can have long-lasting beneficial effects, even after treatment cessation, is also unknown. To address these questions, we first examined the developmental profile for the acquisition of associative learning in a rat model of FXS. Associative memory was tested using a range of behavioral paradigms that rely on an animal's innate tendency to explore novelty. Fmr1 knockout (KO) rats showed a developmental delay in their acquisition of object-place recognition and did not demonstrate object-place-context recognition paradigm at any age tested (up to 23 weeks of age). Treatment of Fmr1 KO rats with lovastatin between 5 and 9 weeks of age, during the normal developmental period that this associative memory capability is established, prevents the emergence of deficits but has no effect in wild-type animals. Moreover, we observe no regression of cognitive performance in the FXS rats over several months after treatment. This restoration of the normal developmental trajectory of cognitive function is associated with the sustained rescue of both synaptic plasticity and altered protein synthesis. The findings provide proof of concept that the impaired emergence of the cognitive repertoire in neurodevelopmental disorders may be prevented by brief, early pharmacological intervention.


Assuntos
Síndrome do Cromossomo X Frágil/fisiopatologia , Síndrome do Cromossomo X Frágil/terapia , Aprendizagem , Animais , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Proteína do X Frágil da Deficiência Intelectual/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Lovastatina/farmacologia , Masculino , Memória/efeitos dos fármacos , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Reconhecimento Psicológico/efeitos dos fármacos , Análise e Desempenho de Tarefas
12.
Neuron ; 65(3): 385-98, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20159451

RESUMO

Alterations in sensory processing constitute prominent symptoms of fragile X syndrome; however, little is known about how disrupted synaptic and circuit development in sensory cortex contributes to these deficits. To investigate how the loss of fragile X mental retardation protein (FMRP) impacts the development of cortical synapses, we examined excitatory thalamocortical synapses in somatosensory cortex during the perinatal critical period in Fmr1 knockout mice. FMRP ablation resulted in dysregulation of glutamatergic signaling maturation. The fraction of silent synapses persisting to later developmental times was increased; there was a temporal delay in the window for synaptic plasticity, while other forms of developmental plasticity were not altered in Fmr1 knockout mice. Our results indicate that FMRP is required for the normal developmental progression of synaptic maturation, and loss of this important RNA binding protein impacts the timing of the critical period for layer IV synaptic plasticity.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Plasticidade Neuronal/fisiologia , Córtex Somatossensorial/citologia , Córtex Somatossensorial/metabolismo , 2-Amino-5-fosfonovalerato/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Guanilato Quinases , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica/métodos , Vias Neurais/crescimento & desenvolvimento , Técnicas de Patch-Clamp/métodos , Receptores de Glutamato/fisiologia , Córtex Somatossensorial/crescimento & desenvolvimento , Córtex Somatossensorial/ultraestrutura , Tálamo/crescimento & desenvolvimento , Fatores de Tempo , Vibrissas/lesões , Vibrissas/inervação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA