Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 54(11): 2611-2631.e8, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34758338

RESUMO

Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.


Assuntos
Hipocampo/metabolismo , Interleucina-6/biossíntese , Exposição Materna , Neurônios/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Sinapses/metabolismo , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Hipocampo/fisiopatologia , Mediadores da Inflamação/metabolismo , Camundongos , Gravidez , Transdução de Sinais , Transmissão Sináptica
2.
Neurobiol Dis ; 193: 106465, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38460800

RESUMO

Children who experienced moderate perinatal asphyxia (MPA) are at risk of developing long lasting subtle cognitive and behavioral deficits, including learning disabilities and emotional problems. The prefrontal cortex (PFC) regulates cognitive flexibility and emotional behavior. Neurons that release serotonin (5-HT) project to the PFC, and compounds modulating 5-HT activity influence emotion and cognition. Whether 5-HT dysregulations contribute to MPA-induced cognitive problems is unknown. We established a MPA mouse model, which displays recognition and spatial memory impairments and dysfunctional cognitive flexibility. We found that 5-HT expression levels, quantified by immunohistochemistry, and 5-HT release, quantified by in vivo microdialysis in awake mice, are reduced in PFC of adult MPA mice. MPA mice also show impaired body temperature regulation following injection of the 5-HT1A receptor agonist 8-OH-DPAT, suggesting the presence of deficits in 5-HT auto-receptor function on raphe neurons. Finally, chronic treatment of adult MPA mice with fluoxetine, an inhibitor of 5-HT reuptake transporter, or the 5-HT1A receptor agonist tandospirone rescues cognitive flexibility and memory impairments. All together, these data demonstrate that the development of 5-HT system function is vulnerable to moderate perinatal asphyxia. 5-HT hypofunction might in turn contribute to long-term cognitive impairment in adulthood, indicating a potential target for pharmacological therapies.


Assuntos
Inibidores Seletivos de Recaptação de Serotonina , Serotonina , Humanos , Criança , Camundongos , Animais , Serotonina/metabolismo , Receptor 5-HT1A de Serotonina , Asfixia , Fluoxetina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Receptores de Serotonina , Cognição , 8-Hidroxi-2-(di-n-propilamino)tetralina/farmacologia , Hipóxia
3.
Mol Psychiatry ; 28(7): 2946-2963, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37131076

RESUMO

While persistence of fear memories is essential for survival, a failure to inhibit fear in response to harmless stimuli is a feature of anxiety disorders. Extinction training only temporarily suppresses fear memory recovery in adults, but it is highly effective in juvenile rodents. Maturation of GABAergic circuits, in particular of parvalbumin-positive (PV+) cells, restricts plasticity in the adult brain, thus reducing PV+ cell maturation could promote the suppression of fear memories following extinction training in adults. Epigenetic modifications such as histone acetylation control gene accessibility for transcription and help couple synaptic activity to changes in gene expression. Histone deacetylase 2 (Hdac2), in particular, restrains both structural and functional synaptic plasticity. However, whether and how Hdac2 controls the maturation of postnatal PV+ cells is not well understood. Here, we show that PV+- cell specific Hdac2 deletion limits spontaneous fear memory recovery in adult mice, while enhancing PV+ cell bouton remodeling and reducing perineuronal net aggregation around PV+ cells in prefrontal cortex and basolateral amygdala. Prefrontal cortex PV+ cells lacking Hdac2, show reduced expression of Acan, a critical perineuronal net component, which is rescued by Hdac2 re-expression. Pharmacological inhibition of Hdac2 before extinction training is sufficient to reduce both spontaneous fear memory recovery and Acan expression in wild-type adult mice, while these effects are occluded in PV+-cell specific Hdac2 conditional knockout mice. Finally, a brief knock-down of Acan expression mediated by intravenous siRNA delivery before extinction training but after fear memory acquisition is sufficient to reduce spontaneous fear recovery in wild-type mice. Altogether, these data suggest that controlled manipulation of PV+ cells by targeting Hdac2 activity, or the expression of its downstream effector Acan, promotes the long-term efficacy of extinction training in adults.


Assuntos
Condicionamento Psicológico , Parvalbuminas , Camundongos , Animais , Parvalbuminas/metabolismo , Regulação para Baixo , Condicionamento Psicológico/fisiologia , Memória/fisiologia , Medo/fisiologia , Camundongos Knockout , Extinção Psicológica/fisiologia
4.
Neurobiol Dis ; 180: 106097, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967064

RESUMO

We review here the neuronal mechanisms that cause seizures in focal epileptic disorders and, specifically, those involving limbic structures that are known to be implicated in human mesial temporal lobe epilepsy. In both epileptic patients and animal models, the initiation of focal seizures - which are most often characterized by a low-voltage fast onset EEG pattern - is presumably dependent on the synchronous firing of GABA-releasing interneurons that, by activating post-synaptic GABAA receptors, cause large increases in extracellular [K+] through the activation of the co-transporter KCC2. A similar mechanism may contribute to seizure maintenance; accordingly, inhibiting KCC2 activity transforms seizure activity into a continuous pattern of short-lasting epileptiform discharges. It has also been found that interactions between different areas of the limbic system modulate seizure occurrence by controlling extracellular [K+] homeostasis. In line with this view, low-frequency electrical or optogenetic activation of limbic networks restrain seizure generation, an effect that may also involve the activation of GABAB receptors and activity-dependent changes in epileptiform synchronization. Overall, these findings highlight the paradoxical role of GABAA signaling in both focal seizure generation and maintenance, emphasize the efficacy of low-frequency activation in abating seizures, and provide experimental evidence explaining the poor efficacy of antiepileptic drugs designed to augment GABAergic function in controlling seizures in focal epileptic disorders.


Assuntos
Epilepsias Parciais , Simportadores , Animais , Humanos , Ligantes , Convulsões , Receptores de GABA-A , Ácido gama-Aminobutírico
5.
Brain ; 145(2): 754-769, 2022 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-34791091

RESUMO

Amongst the numerous genes associated with intellectual disability, SYNGAP1 stands out for its frequency and penetrance of loss-of-function variants found in patients, as well as the wide range of co-morbid disorders associated with its mutation. Most studies exploring the pathophysiological alterations caused by Syngap1 haploinsufficiency in mouse models have focused on cognitive problems and epilepsy; however, whether and to what extent sensory perception and processing are altered by Syngap1 haploinsufficiency is less clear. By performing EEG recordings in awake mice, we identified specific alterations in multiple aspects of auditory and visual processing, including increased baseline gamma oscillation power, increased theta/gamma phase amplitude coupling following stimulus presentation and abnormal neural entrainment in response to different sensory modality-specific frequencies. We also report lack of habituation to repetitive auditory stimuli and abnormal deviant sound detection. Interestingly, we found that most of these alterations are present in human patients as well, thus making them strong candidates as translational biomarkers of sensory-processing alterations associated with SYNGAP1/Syngap1 haploinsufficiency.


Assuntos
Haploinsuficiência , Deficiência Intelectual , Animais , Biomarcadores , Eletroencefalografia , Haploinsuficiência/genética , Humanos , Deficiência Intelectual/genética , Camundongos , Percepção , Proteínas Ativadoras de ras GTPase/genética
6.
J Neurosci ; 41(43): 8876-8886, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34503995

RESUMO

Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood.SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.


Assuntos
Córtex Cerebral/metabolismo , Neurônios/metabolismo , Parvalbuminas/biossíntese , Fatores de Transcrição SOXD/biossíntese , Sinapses/metabolismo , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Parvalbuminas/genética , Fatores de Transcrição SOXD/genética , Sinapses/genética
7.
Cereb Cortex ; 30(1): 256-268, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31038696

RESUMO

Methyl-CpG-binding protein 2 (MeCP2) mutations are the primary cause of Rett syndrome, a severe neurodevelopmental disorder. Cortical parvalbumin GABAergic interneurons (PV) make exuberant somatic connections onto pyramidal cells in the visual cortex of Mecp2-deficient mice, which contributes to silencing neuronal cortical circuits. This phenotype can be rescued independently of Mecp2 by environmental, pharmacological, and genetic manipulation. It remains unknown how Mecp2 mutation can result in abnormal inhibitory circuit refinement. In the present manuscript, we examined the development of GABAergic circuits in the primary visual cortex of Mecp2-deficient mice. We identified that PV circuits were the only GABAergic interneurons to be upregulated, while other interneurons were downregulated. Acceleration of PV cell maturation was accompanied by increased PV cells engulfment by perineuronal nets (PNNs) and by an increase of PV cellular and PNN structural complexity. Interestingly, selective deletion of Mecp2 from PV cells was sufficient to drive increased structure complexity of PNN. Moreover, the accelerated PV and PNN maturation was recapitulated in organotypic cultures. Our results identify a specific timeline of disruption of GABAergic circuits in the absence of Mecp2, indicating a possible cell-autonomous role of MeCP2 in the formation of PV cellular arbors and PNN structures in the visual cortex.


Assuntos
Neurônios GABAérgicos/fisiologia , Proteína 2 de Ligação a Metil-CpG/fisiologia , Parvalbuminas/fisiologia , Córtex Visual/crescimento & desenvolvimento , Animais , Neurônios GABAérgicos/citologia , Interneurônios/citologia , Interneurônios/fisiologia , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/citologia , Vias Neurais/crescimento & desenvolvimento , Córtex Visual/citologia
8.
J Neurosci ; 39(23): 4489-4510, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-30936240

RESUMO

By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.


Assuntos
Envelhecimento/fisiologia , Interneurônios/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Maturidade Sexual/fisiologia , Córtex Visual/crescimento & desenvolvimento , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Conectoma , Potenciais Evocados Visuais , Feminino , Neurônios GABAérgicos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/química , Interneurônios/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Parvalbuminas/análise , Precursores de Proteínas/farmacologia , Distribuição Aleatória , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Sinapses/fisiologia , Visão Monocular/fisiologia , Córtex Visual/citologia , Córtex Visual/metabolismo
9.
Hum Mol Genet ; 26(12): 2307-2320, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28369449

RESUMO

Scribble1 (Scrib1) is a tumor suppressor gene that has long been established as an essential component of apicobasal polarity (ABP). In mouse models, mutations in Scrib1 cause a severe form of neural tube defects (NTDs) as a result of a defective planar cell polarity (PCP) signaling. In this study, we dissected the role of Scrib1 in the pathogenesis of NTDs in its mouse mutant Circletail (Crc), in cell lines and in a human NTD cohort. While there were no obvious defects in ABP in the Scrib1Crc/Crc neuroepihelial cells, we identified an abnormal localization of the apical protein Par-3 and of the PCP protein Vangl2. These results were concordant with those obtained following a partial knockdown of Scrib1 in MDCK II cells. Par-3 was able to rescue the localization defect of Vangl1 (paralog of Vangl2) caused by partial knockdown of Scrib1 suggesting that Scrib1 exerts its effect on Vangl1 localization indirectly through Par-3. This conclusion is supported by our findings of an apical enrichment of Vangl1 following a partial knockdown of Par-3. Re-sequencing analysis of SCRIB1 in 473 NTD patients led to the identification of 5 rare heterozygous missense mutations that were predicted to be pathogenic. Two of these mutations, p.Gly263Ser and p.Gln808His, and 2 mouse NTD mutations, p.Ile285Lys and p.Glu814Gly, affected Scrib1 membrane localization and its modulating role of Par-3 and Vangl1 localization. Our study demonstrates an important role of Scrib1 in the pathogenesis of NTDs through its mediating effect of Par-3 and Vangl1/2 localization and most likely independently of ABP.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Polaridade Celular/genética , Pré-Escolar , Feminino , Heterozigoto , Humanos , Lactente , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Mutação de Sentido Incorreto , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo
10.
Cereb Cortex ; 28(11): 4049-4062, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30169756

RESUMO

KCC2 is the major chloride extruder in neurons. The spatiotemporal regulation of KCC2 expression orchestrates the developmental shift towards inhibitory GABAergic drive and the formation of glutamatergic synapses. Whether KCC2's role in synapse formation is similar in different brain regions is unknown. First, we found that KCC2 subcellular localization, but not overall KCC2 expression levels, differed between cortex and hippocampus during the first postnatal week. We performed site-specific in utero electroporation of KCC2 cDNA to target either hippocampal CA1 or somatosensory cortical pyramidal neurons. We found that a premature expression of KCC2 significantly decreased spine density in CA1 neurons, while it had the opposite effect in cortical neurons. These effects were cell autonomous, because single-cell biolistic overexpression of KCC2 in hippocampal and cortical organotypic cultures also induced a reduction and an increase of dendritic spine density, respectively. In addition, we found that the effects of its premature expression on spine density were dependent on BDNF levels. Finally, we showed that the effects of KCC2 on dendritic spine were dependent on its chloride transporter function in the hippocampus, contrary to what was observed in cortex. Altogether, these results demonstrate that KCC2 regulation of dendritic spine development, and its underlying mechanisms, are brain-region specific.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Região CA1 Hipocampal/crescimento & desenvolvimento , Espinhas Dendríticas/fisiologia , Córtex Somatossensorial/crescimento & desenvolvimento , Simportadores/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA1 Hipocampal/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células Piramidais/fisiologia , Ratos Sprague-Dawley , Simportadores/metabolismo , Cotransportadores de K e Cl-
11.
Mol Cell Neurosci ; 79: 81-92, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28088609

RESUMO

Minipump infusions into visual cortex in vivo at the onset of the critical period have revealed that the proinflammatory cytokine leukemia inhibitory factor (LIF) delays the maturation of thalamocortical projection neurons of the lateral geniculate nucleus, and tecto-thalamic projection neurons of the superior colliculus, and cortical layer IV spiny stellates and layer VI pyramidal neurons. Here, we report that P12-20 LIF infusion inhibits somatic maturation of pyramidal neurons and of all interneuron types in vivo. Likewise, DIV 12-20 LIF treatment in organotypic cultures prevents somatic growth GABA-ergic neurons. Further, while NPY expression is increased in the LIF-infused hemispheres, the expression of parvalbumin mRNA and protein, Kv3.1 mRNA, calbindin D-28k protein, and GAD-65 mRNA, but not of GAD-67 mRNA or calretinin protein is substantially reduced. Also, LIF treatment decreases parvalbumin, Kv3.1, Kv3.2 and GAD-65, but not GAD-67 mRNA expression in OTC. Developing cortical neurons are known to depend on neurotrophins. Indeed, LIF alters neurotrophin mRNA expression, and prevents the growth promoting action of neurotophin-4 in GABA-ergic neurons. The results imply that LIF, by altering neurotrophin expression and/or signaling, could counteract neurotrophin-dependent growth and neurochemical differentiation of cortical neurons.


Assuntos
Fator Inibidor de Leucemia/farmacologia , Neurogênese/efeitos dos fármacos , Córtex Visual/efeitos dos fármacos , Animais , Células Cultivadas , Feminino , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Interneurônios/citologia , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Ratos Long-Evans , Córtex Visual/citologia , Córtex Visual/crescimento & desenvolvimento
12.
Neurobiol Dis ; 91: 10-20, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26875662

RESUMO

Atypical febrile seizures are considered a risk factor for epilepsy onset and cognitive impairments later in life. Patients with temporal lobe epilepsy and a history of atypical febrile seizures often carry a cortical malformation. This association has led to the hypothesis that the presence of a cortical dysplasia exacerbates febrile seizures in infancy, in turn increasing the risk for neurological sequelae. The mechanisms linking these events are currently poorly understood. Potassium-chloride cotransporter KCC2 affects several aspects of neuronal circuit development and function, by modulating GABAergic transmission and excitatory synapse formation. Recent data suggest that KCC2 downregulation contributes to seizure generation in the epileptic adult brain, but its role in the developing brain is still controversial. In a rodent model of atypical febrile seizures, combining a cortical dysplasia and hyperthermia-induced seizures (LHS rats), we found a premature and sustained increase in KCC2 protein levels, accompanied by a negative shift of the reversal potential of GABA. In parallel, we observed a significant reduction in dendritic spine size and mEPSC amplitude in CA1 pyramidal neurons, accompanied by spatial memory deficits. To investigate whether KCC2 premature overexpression plays a role in seizure susceptibility and synaptic alterations, we reduced KCC2 expression selectively in hippocampal pyramidal neurons by in utero electroporation of shRNA. Remarkably, KCC2 shRNA-electroporated LHS rats show reduced hyperthermia-induced seizure susceptibility, while dendritic spine size deficits were rescued. Our findings demonstrate that KCC2 overexpression in a compromised developing brain increases febrile seizure susceptibility and contribute to dendritic spine alterations.


Assuntos
Encéfalo/metabolismo , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Convulsões Febris/patologia , Simportadores/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Suscetibilidade a Doenças/metabolismo , Epilepsia/fisiopatologia , Transtornos da Memória/metabolismo , Neurogênese/fisiologia , Células Piramidais/metabolismo , Ratos Sprague-Dawley , Convulsões Febris/metabolismo , Convulsões Febris/fisiopatologia , Cotransportadores de K e Cl-
13.
J Neurosci ; 33(14): 5957-68, 2013 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-23554477

RESUMO

GABAergic basket interneurons form perisomatic synapses, which are essential for regulating neural networks, and their alterations are linked to various cognitive dysfunction. Maturation of basket synapses in postnatal cortex is activity dependent. In particular, activity-dependent downregulation of polysialiac acid carried by the neural cell adhesion molecule (NCAM) regulates the timing of their maturation. Whether and how NCAM per se affects GABAergic synapse development is unknown. Using single-cell genetics to knock out NCAM in individual basket interneurons in mouse cortical slice cultures, at specific developmental time periods, we found that NCAM loss during perisomatic synapse formation impairs the process of basket cell axonal branching and bouton formation. However, loss of NCAM once the synapses are already formed did not show any effect. We further show that NCAM120 and NCAM140, but not the NCAM180 isoform, rescue the phenotype. Finally, we demonstrate that a dominant-negative form of Fyn kinase mimics, whereas a constitutively active form of Fyn kinase rescues, the effects of NCAM knockdown. Altogether, our data suggest that NCAM120/NCAM140-mediated Fyn activation promotes GABAergic synapse maturation in postnatal cortex.


Assuntos
Córtex Cerebral/citologia , Interneurônios/fisiologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Sinapses/fisiologia , Ácido gama-Aminobutírico/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , Moléculas de Adesão de Célula Nervosa/genética , Inibição Neural/fisiologia , Técnicas de Cultura de Órgãos , Fosfopiruvato Hidratase/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fyn/genética , Estatísticas não Paramétricas , Sinapses/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
14.
Hum Mol Genet ; 21(10): 2211-8, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22337953

RESUMO

Spinocerebellar ataxia type 3 is caused by the expansion of the coding CAG repeat in the ATXN3 gene. Interestingly, a -1 bp frameshift occurring within an (exp)CAG repeat would henceforth lead to translation from a GCA frame, generating polyalanine stretches instead of polyglutamine. Our results show that transgenic expression of (exp)CAG ATXN3 led to -1 frameshifting events, which have deleterious effects in Drosophila and mammalian neurons. Conversely, transgenic expression of polyglutamine-encoding (exp)CAA ATXN3 was not toxic. Furthermore, (exp)CAG ATXN3 mRNA does not contribute per se to the toxicity observed in our models. Our observations indicate that expanded polyglutamine tracts in Drosophila and mouse neurons are insufficient for the development of a phenotype. Hence, we propose that -1 ribosomal frameshifting contributes to the toxicity associated with (exp)CAG repeats.


Assuntos
Drosophila/genética , Mudança da Fase de Leitura do Gene Ribossômico , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Ataxina-3 , Drosophila/metabolismo , Imuno-Histoquímica , Doença de Machado-Joseph/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/química , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Transfecção , Expansão das Repetições de Trinucleotídeos
15.
Curr Res Neurobiol ; 6: 100131, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38812499

RESUMO

Catamenial epilepsy, defined as a periodicity of seizure exacerbation during the menstrual cycle, affects up to 70 % of epileptic women. Seizures in these patients are often non-responsive to medication; however, our understanding of the relation between menstrual cycle and seizure generation (i.e. ictogenesis) remains limited. We employed here field potential recordings in the in vitro 4-aminopyridine model of epileptiform synchronization in female mice (P60-P130) and found that: (i) the estrous phase favors ictal activity in the entorhinal cortex; (ii) these ictal discharges display an onset pattern characterised by the presence of chirps that are thought to mirror synchronous interneuron firing; and (iii) blocking estrogen receptor ß-mediated signaling reduces ictal discharge duration. Our findings indicate that the duration of 4AP-induced ictal discharges, in vitro, increases during the estrous phase, which corresponds to the human peri-ovulatory period. We propose that these effects are caused by the presumptive enhancement of interneuron excitability due to increased estrogen receptor ß-mediated signaling.

16.
J Neurosci ; 32(3): 911-8, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-22262889

RESUMO

Neural activity guides the patterning of neuron synaptic territory in the developing nervous system. Evidence supporting this hypothesis comes from numerous studies on projection neurons in neuromuscular and visual systems. It is unknown whether the innervation field of GABAergic interneurons, which forms local dense innervations, follows similar rules. Cortical basket cells innervate hundreds of pyramidal cell somata and proximal dendrites. Thanks to this connectivity pattern, they can tightly control neural excitability and synchronization. Here we show that reducing excitation, and thus neurotransmitter release, in mouse cortical single basket cells in slice cultures decreases the number of innervated cells without changing the pattern of perisomatic innervation, both at the peak and after the proliferation phase of perisomatic synapse formation. Conversely, suppressing neurotransmitter release in single basket cells can have completely opposite effects depending on the developmental stage. Our results reveal a remarkably specific and age-dependent role of neural activity and neurotransmission levels in the establishment of the synaptic territory of cortical GABAergic cells.


Assuntos
Envelhecimento/fisiologia , Córtex Cerebral/citologia , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Inibição Neural/fisiologia , Sinapses/fisiologia , Envelhecimento/genética , Análise de Variância , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Proteína de Ligação a CREB/metabolismo , Córtex Cerebral/efeitos dos fármacos , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Interneurônios/citologia , Interneurônios/efeitos dos fármacos , Masculino , Metaloendopeptidases/farmacologia , Camundongos , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neuropeptídeos/química , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Peptídeos/farmacologia , Receptores de Neuropeptídeos/genética , Sinapses/efeitos dos fármacos , Sinapses/genética , Toxina Tetânica/farmacologia , Transfecção
17.
J Neurosci ; 32(1): 331-43, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22219294

RESUMO

Accumulating evidence indicates that GABA acts beyond inhibitory synaptic transmission and regulates the development of inhibitory synapses in the vertebrate brain, but the underlying cellular mechanism is not well understood. We have combined live imaging of cortical GABAergic axons across time scales from minutes to days with single-cell genetic manipulation of GABA release to examine its role in distinct steps of inhibitory synapse formation in the mouse neocortex. We have shown previously, by genetic knockdown of GABA synthesis in developing interneurons, that GABA signaling promotes the maturation of inhibitory synapses and axons. Here we found that a complete blockade of GABA release in basket interneurons resulted in an opposite effect, a cell-autonomous increase in axon and bouton density with apparently normal synapse structures. These results not only demonstrate that GABA is unnecessary for synapse formation per se but also uncover a novel facet of GABA in regulating synapse elimination and axon pruning. Live imaging revealed that developing GABAergic axons form a large number of transient boutons, but only a subset was stabilized. Release blockade led to significantly increased bouton stability and filopodia density, increased axon branch extension, and decreased branch retraction. Our results suggest that a major component of GABA function in synapse development is transmission-mediated elimination of subsets of nascent contacts. Therefore, GABA may regulate activity-dependent inhibitory synapse formation by coordinately eliminating certain nascent contacts while promoting the maturation of other nascent synapses.


Assuntos
Axônios/fisiologia , Diferenciação Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Interneurônios/fisiologia , Sinapses/fisiologia , Ácido gama-Aminobutírico/deficiência , Animais , Animais Recém-Nascidos , Axônios/ultraestrutura , Córtex Cerebral/citologia , Feminino , Interneurônios/citologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Inibição Neural/fisiologia , Neurogênese/fisiologia , Técnicas de Cultura de Órgãos , Sinapses/ultraestrutura , Ácido gama-Aminobutírico/biossíntese , Ácido gama-Aminobutírico/genética
18.
Hum Mutat ; 34(2): 385-94, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23161826

RESUMO

De novo mutations in SYNGAP1, which codes for a RAS/RAP GTP-activating protein, cause nonsyndromic intellectual disability (NSID). All disease-causing point mutations identified until now in SYNGAP1 are truncating, raising the possibility of an association between this type of mutations and NSID. Here, we report the identification of the first pathogenic missense mutations (c.1084T>C [p.W362R], c.1685C>T [p.P562L]) and three novel truncating mutations (c.283dupC [p.H95PfsX5], c.2212_2213del [p.S738X], and (c.2184del [p.N729TfsX31]) in SYNGAP1 in patients with NSID. A subset of these patients also showed ataxia, autism, and a specific form of generalized epilepsy that can be refractory to treatment. All of these mutations occurred de novo, except c.283dupC, which was inherited from a father who is a mosaic. Biolistic transfection of wild-type SYNGAP1 in pyramidal cells from cortical organotypic cultures significantly reduced activity-dependent phosphorylated extracellular signal-regulated kinase (pERK) levels. In contrast, constructs expressing p.W362R, p.P562L, or the previously described p.R579X had no significant effect on pERK levels. These experiments suggest that the de novo missense mutations, p.R579X, and possibly all the other truncating mutations in SYNGAP1 result in a loss of its function. Moreover, our study confirms the involvement of SYNGAP1 in autism while providing novel insight into the epileptic manifestations associated with its disruption.


Assuntos
Transtorno Autístico/genética , Epilepsia/genética , Haploinsuficiência , Deficiência Intelectual/genética , Proteínas Ativadoras de ras GTPase/genética , Adolescente , Sequência de Aminoácidos , Transtorno Autístico/fisiopatologia , Western Blotting , Criança , Pré-Escolar , Clonagem Molecular , Epilepsia/fisiopatologia , Exoma , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Células HEK293 , Humanos , Deficiência Intelectual/fisiopatologia , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Fenótipo , Fosforilação , Conformação Proteica , Análise de Sequência de DNA , Transfecção , Proteínas Ativadoras de ras GTPase/metabolismo
19.
PLoS Genet ; 6(9): e1001118, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862357

RESUMO

Tay-Sachs disease is a severe lysosomal disorder caused by mutations in the HexA gene coding for the α-subunit of lysosomal ß-hexosaminidase A, which converts G(M2) to G(M3) ganglioside. Hexa(-/-) mice, depleted of ß-hexosaminidase A, remain asymptomatic to 1 year of age, because they catabolise G(M2) ganglioside via a lysosomal sialidase into glycolipid G(A2), which is further processed by ß-hexosaminidase B to lactosyl-ceramide, thereby bypassing the ß-hexosaminidase A defect. Since this bypass is not effective in humans, infantile Tay-Sachs disease is fatal in the first years of life. Previously, we identified a novel ganglioside metabolizing sialidase, Neu4, abundantly expressed in mouse brain neurons. Now we demonstrate that mice with targeted disruption of both Neu4 and Hexa genes (Neu4(-/-);Hexa(-/-)) show epileptic seizures with 40% penetrance correlating with polyspike discharges on the cortical electrodes of the electroencephalogram. Single knockout Hexa(-/-) or Neu4(-/-) siblings do not show such symptoms. Further, double-knockout but not single-knockout mice have multiple degenerating neurons in the cortex and hippocampus and multiple layers of cortical neurons accumulating G(M2) ganglioside. Together, our data suggest that the Neu4 block exacerbates the disease in Hexa(-/-) mice, indicating that Neu4 is a modifier gene in the mouse model of Tay-Sachs disease, reducing the disease severity through the metabolic bypass. However, while disease severity in the double mutant is increased, it is not profound suggesting that Neu4 is not the only sialidase contributing to the metabolic bypass in Hexa(-/-) mice.


Assuntos
Epilepsia/enzimologia , Epilepsia/patologia , Lisossomos/enzimologia , Neuraminidase/deficiência , Neurônios/enzimologia , Neurônios/patologia , Cadeia alfa da beta-Hexosaminidase/metabolismo , Animais , Comportamento Animal , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Córtex Cerebral/ultraestrutura , Eletroencefalografia , Epilepsia/fisiopatologia , Gangliosídeo G(M2)/metabolismo , Técnicas de Inativação de Genes , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Aprendizagem/fisiologia , Lisossomos/patologia , Lisossomos/ultraestrutura , Camundongos , Atividade Motora/fisiologia , Neuraminidase/metabolismo , Neurônios/ultraestrutura
20.
Biol Psychiatry ; 94(4): 310-321, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37120061

RESUMO

BACKGROUND: Parvalbumin (PV)-positive GABAergic (gamma-aminobutyric acidergic) cells provide robust perisomatic inhibition to neighboring pyramidal neurons and regulate brain oscillations. Alterations in PV interneuron connectivity and function in the medial prefrontal cortex have been consistently reported in psychiatric disorders associated with cognitive rigidity, suggesting that PV cell deficits could be a core cellular phenotype in these disorders. The p75 neurotrophin receptor (p75NTR) regulates the time course of PV cell maturation in a cell-autonomous fashion. Whether p75NTR expression during postnatal development affects adult prefrontal PV cell connectivity and cognitive function is unknown. METHODS: We generated transgenic mice with conditional knockout of p75NTR in postnatal PV cells. We analyzed PV cell connectivity and recruitment following a tail pinch by immunolabeling and confocal imaging in naïve mice or following p75NTR re-expression in preadolescent or postadolescent mice using Cre-dependent viral vectors. Cognitive flexibility was evaluated using behavioral tests. RESULTS: PV cell-specific p75NTR deletion increased both PV cell synapse density and proportion of PV cells surrounded by perineuronal nets, a marker of mature PV cells, in adult medial prefrontal cortex, but not visual cortex. Both phenotypes were rescued by viral-mediated reintroduction of p75NTR in preadolescent, but not postadolescent, medial prefrontal cortex. Prefrontal cortical PV cells failed to upregulate c-Fos following a tail-pinch stimulation in adult conditional knockout mice. Finally, conditional knockout mice showed impaired fear memory extinction learning as well as deficits in an attention set-shifting task. CONCLUSIONS: These findings suggest that p75NTR expression in adolescent PV cells contributes to the fine-tuning of their connectivity and promotes cognitive flexibility in adulthood.


Assuntos
Parvalbuminas , Receptor de Fator de Crescimento Neural , Animais , Camundongos , Cognição , Interneurônios/fisiologia , Camundongos Knockout , Camundongos Transgênicos , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA