Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
eNeuro ; 10(5)2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37072176

RESUMO

SYNGAP1 haploinsufficiency in humans causes intellectual disability (ID). SYNGAP1 is highly expressed in cortical excitatory neurons and, reducing its expression in mice accelerates the maturation of excitatory synapses during sensitive developmental periods, restricts the critical period window for plasticity, and impairs cognition. However, its specific role in interneurons remains largely undetermined. In this study, we investigated the effects of conditional Syngap1 disruption in medial ganglionic eminence (MGE)-derived interneurons on hippocampal interneuron firing properties and excitatory synaptic inputs, as well as on pyramidal cell synaptic inhibition and synaptic integration. We show that conditional Syngap1 disruption in MGE-derived interneurons results in cell-specific impairment of firing properties of hippocampal Nkx2.1 fast-spiking interneurons, with enhancement of their AMPA receptor (AMPAR)-mediated excitatory synaptic inputs but compromised short-term plasticity. In contrast, regular-spiking Nkx2.1 interneurons are largely unaffected. These changes are associated with impaired pyramidal cell synaptic inhibition and enhanced summation of excitatory responses. Unexpectedly, we found that the Syngap1flox allele used in this study contains inverted loxP sites and that its targeted recombination in MGE-derived interneurons induces some cell loss during embryonic development and the reversible inversion of the sequence flanked by the loxP sites in postmitotic cells. Together, these results suggest that Syngap1 plays a role in cell-specific regulation of hippocampal interneuron function and inhibition of pyramidal cells in mice. However, because of our finding that the Syngap1flox allele used in this study contains inverted loxP sites, it will be important to further investigate interneuron function using a different Syngap1 conditional allele.


Assuntos
Interneurônios , Células Piramidais , Humanos , Camundongos , Animais , Camundongos Transgênicos , Interneurônios/fisiologia , Células Piramidais/fisiologia , Hipocampo/metabolismo , Recombinação Genética , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
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.
Neurobiol Dis ; 160: 105535, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34673150

RESUMO

BACKGROUND: Recent studies report infiltration of peripheral blood mononuclear cells (PBMCs) into the central nervous system (CNS) in epileptic disorders, suggestive of a potential contribution of PBMC extravasation to the generation of seizures. Nevertheless, the underlying mechanisms involved in PBMC infiltrates promoting neuronal predisposition to ictogenesis remain unclear. Therefore, we developed an in vitro model mimicking infiltration of activated PBMCs into the brain in order to investigate potential transduction of inflammatory signals from PBMCs to the CNS. METHODS: To establish our model, we first extracted PBMCs from rat spleen, then, immunologically primed PBMCs with lipopolysaccharide (LPS), followed by further activation with nigericin. Thereafter, we co-cultured these activated PBMCs with organotypic cortico-hippocampal brain slice cultures (OCHSCs) derived from the same rat, and compared PBMC-OCHSC co-cultures to OCHSCs exposed to PBMCs in the culture media. We further targeted a potential molecular pathway underlying transduction of peripheral inflammation to OCHSCs by incubating OCHSCs with the Caspase-1 inhibitor VX-765 prior to co-culturing PBMCs with OCHSCs. After 24 h, we analyzed inflammation markers in the cortex and the hippocampus using semiquantitative immunofluorescence. In addition, we analyzed neuronal activity by whole-cell patch-clamp recordings in cortical layer II/III and hippocampal CA1 pyramidal neurons. RESULTS: In the cortex, co-culturing immunoreactive PBMCs treated with LPS + nigericin on top of OCHSCs upregulated inflammatory markers and enhanced neuronal excitation. In contrast, no excitability changes were detected after adding primed PBMCs (i.e. treated with LPS only), to OCHSCs. Strikingly, in the hippocampus, both immunoreactive and primed PBMCs elicited similar pro-inflammatory and pro-excitatory effects. However, when immunoreactive and primed PBMCs were cultured in the media separately from OCHSCs, only immunoreactive PBMCs gave rise to neuroinflammation and hyperexcitability in the hippocampus, whereas primed PBMCs failed to produce any significant changes. Finally, VX-765 application to OCHSCs, co-cultured with either immunoreactive or primed PBMCs, prevented neuroinflammation and hippocampal hyperexcitability in OCHSCs. CONCLUSIONS: Our study shows a higher susceptibility of the hippocampus to peripheral inflammation as compared to the cortex, mediated via Caspase-1-dependent signaling pathways. Thus, our findings suggest that Caspase-1 inhibition may potentially provide therapeutic benefits during hippocampal neuroinflammation and hyperexcitability secondary to peripheral innate immunity.


Assuntos
Caspase 1/metabolismo , Hipocampo/metabolismo , Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Neurônios/metabolismo , Animais , Inflamação/induzido quimicamente , Lipopolissacarídeos , Ratos , Ratos Sprague-Dawley
7.
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
8.
Nat Commun ; 12(1): 3653, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34135323

RESUMO

The Mechanistic Target Of Rapamycin Complex 1 (mTORC1) pathway controls several aspects of neuronal development. Mutations in regulators of mTORC1, such as Tsc1 and Tsc2, lead to neurodevelopmental disorders associated with autism, intellectual disabilities and epilepsy. The correct development of inhibitory interneurons is crucial for functional circuits. In particular, the axonal arborisation and synapse density of parvalbumin (PV)-positive GABAergic interneurons change in the postnatal brain. How and whether mTORC1 signaling affects PV cell development is unknown. Here, we show that Tsc1 haploinsufficiency causes a premature increase in terminal axonal branching and bouton density formed by mutant PV cells, followed by a loss of perisomatic innervation in adult mice. PV cell-restricted Tsc1 haploinsufficient and knockout mice show deficits in social behavior. Finally, we identify a sensitive period during the third postnatal week during which treatment with the mTOR inhibitor Rapamycin rescues deficits in both PV cell innervation and social behavior in adult conditional haploinsufficient mice. Our findings reveal a role of mTORC1 signaling in the regulation of the developmental time course and maintenance of cortical PV cell connectivity and support a mechanistic basis for the targeted rescue of autism-related behaviors in disorders associated with deregulated mTORC1 signaling.


Assuntos
Interneurônios/patologia , Parvalbuminas/metabolismo , Comportamento Social , Proteína 1 do Complexo Esclerose Tuberosa/deficiência , Animais , Autofagia , Axônios/efeitos dos fármacos , Axônios/patologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Mutação , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Sirolimo/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/patologia , Fatores de Tempo , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo
9.
Handb Clin Neurol ; 173: 43-53, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32958189

RESUMO

Optimal brain function critically hinges on the remarkably precise interconnections made among millions of neurons. These specialized interconnected neuronal junctions, termed synapses, are used for neuronal communication, whence the presynaptic neurons releases a specific neurotransmitter, which then binds to the appropriate protein receptor on the membrane of the postsynaptic neuron, activating and eliciting a response in this connected neuron. In this chapter, we discuss how synapses form and are modified as the brain matures. Genetic programs control most of the wiring in the brain, from allowing axons to choose where to target their synapses, to determining synapse identity. However, the final map of neuronal connectivity in the brain crucially relies on incoming sensory information during early childhood to strengthen and refine the preexisting synapses thus allowing both nature and nurture to shape the final structure and function of the nervous system (Fig. 5.1). Finally, we discuss how advances in the knowledge of basic mechanisms governing synapse formation and plasticity can shed light on the pathophysiology of neurodevelopmental disorders.


Assuntos
Plasticidade Neuronal , Sinapses , Axônios , Humanos , Neurogênese , Neurônios
10.
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
11.
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
12.
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
13.
Nat Commun ; 7: 13340, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27827368

RESUMO

Haploinsufficiency of the SYNGAP1 gene, which codes for a Ras GTPase-activating protein, impairs cognition both in humans and in mice. Decrease of Syngap1 in mice has been previously shown to cause cognitive deficits at least in part by inducing alterations in glutamatergic neurotransmission and premature maturation of excitatory connections. Whether Syngap1 plays a role in the development of cortical GABAergic connectivity and function remains unclear. Here, we show that Syngap1 haploinsufficiency significantly reduces the formation of perisomatic innervations by parvalbumin-positive basket cells, a major population of GABAergic neurons, in a cell-autonomous manner. We further show that Syngap1 haploinsufficiency in GABAergic cells derived from the medial ganglionic eminence impairs their connectivity, reduces inhibitory synaptic activity and cortical gamma oscillation power, and causes cognitive deficits. Our results indicate that Syngap1 plays a critical role in GABAergic circuit function and further suggest that Syngap1 haploinsufficiency in GABAergic circuits may contribute to cognitive deficits.


Assuntos
Transtornos Cognitivos/genética , Cognição/fisiologia , Neurônios GABAérgicos/fisiologia , Sinapses/fisiologia , Proteínas Ativadoras de ras GTPase/fisiologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Haploinsuficiência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cultura Primária de Células , Transmissão Sináptica/fisiologia , Proteínas Ativadoras de ras GTPase/genética
14.
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
15.
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
16.
Front Psychiatry ; 3: 51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666213

RESUMO

GABAergic interneurons control neuronal excitability, integration, and plasticity. Further, they regulate the generation of temporal synchrony and oscillatory behavior among networks of pyramidal neurons. Such oscillations within and across neural systems are believed to serve various complex functions, such as perception, movement initiation, and memory. Alterations in the development of GABAergic circuits have been implicated in various brain diseases with neurodevelopmental origin. Here, we highlight recent studies suggesting a role for alterations of GABA transmission in the pathophysiology of two neurodevelopmental diseases, schizophrenia, and autism. We further discuss how manipulations of GABA signaling may be used for novel therapeutic interventions.

17.
Neural Plast ; 2011: 734231, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21826279

RESUMO

GABAergic interneurons are critical for the normal function and development of neural circuits, and their dysfunction is implicated in a large number of neurodevelopmental disorders. Experience and activity-dependent mechanisms play an important role in GABAergic circuit development, also recent studies involve a number of molecular players involved in the process. Emphasizing the molecular mechanisms of GABAergic synapse formation, in particular basket cell perisomatic synapses, this paper draws attention to the links between critical period plasticity, GABAergic synapse maturation, and the consequences of its dysfunction on the development of the nervous system.


Assuntos
Deficiências do Desenvolvimento/metabolismo , Doenças do Sistema Nervoso/metabolismo , Plasticidade Neuronal , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Período Crítico Psicológico , Deficiências do Desenvolvimento/fisiopatologia , Humanos , Rede Nervosa/crescimento & desenvolvimento , Doenças do Sistema Nervoso/fisiopatologia
18.
Neurobiol Dis ; 43(2): 312-21, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21406232

RESUMO

Clinical evidence suggests that febrile status epilepticus (SE) in children can lead to acute hippocampal injury and subsequent temporal lobe epilepsy. The contribution of febrile SE to the mechanisms underlying temporal lobe epilepsy are however poorly understood. A rat model of temporal lobe epilepsy following hyperthermic SE was previously established in our laboratory, wherein a focal cortical lesion induced at postnatal day 1 (P1), followed by a hyperthermic SE (more than 30 min) at P10, leads to hippocampal atrophy at P22 (dual pathology model) and spontaneous recurrent seizures (SRS) with mild visuospatial memory deficits in adult rats. The goal of this study was to identify the long term electrophysiological, anatomical and molecular changes in this model. Following hyperthermic SE, all cortically lesioned pups developed progressive SRS as adults, characterized by the onset of highly rhythmic activity in the hippocampus. A reduction of hippocampal volume on the side of the lesion preceded the SRS and was associated with a loss of hippocampal neurons, a marked decrease in pyramidal cell spine density, an increase in the hippocampal levels of NMDA receptor NR2A subunit, but no significant change in GABA receptors. These findings suggest that febrile SE in the abnormal brain leads to hippocampal injury that is followed by progressive network reorganization and molecular changes that contribute to the epileptogenesis as well as the observed memory deficits.


Assuntos
Epilepsia do Lobo Temporal/complicações , Epilepsia do Lobo Temporal/patologia , Convulsões Febris/patologia , Doença Aguda , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/fisiopatologia , Feminino , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Ratos , Ratos Sprague-Dawley , Convulsões Febris/complicações , Convulsões Febris/fisiopatologia , Fatores de Tempo
19.
Artigo em Inglês | MEDLINE | ID: mdl-20126518

RESUMO

The conditional expression of transgenes at high levels in sparse and specific populations of neurons is important for high-resolution optogenetic analyses of neuronal circuits. We explored two complementary methods, viral gene delivery and the iTet-Off system, to express transgenes in the brain of zebrafish. High-level gene expression in neurons was achieved by Sindbis and Rabies viruses. The Tet system produced strong and specific gene expression that could be modulated conveniently by doxycycline. Moreover, transgenic lines showed expression in distinct, sparse and stable populations of neurons that appeared to be subsets of the neurons targeted by the promoter driving the Tet-activator. The Tet system therefore provides the opportunity to generate libraries of diverse expression patterns similar to gene trap approaches or the thy-1 promoter in mice, but with the additional possibility to pre-select cell types of interest. In transgenic lines expressing channelrhodopsin-2, action potential firing could be precisely controlled by two-photon stimulation at low laser power, presumably because the expression levels of the Tet-controlled genes were high even in adults. In channelrhodopsin-2-expressing larvae, optical stimulation with a single blue LED evoked distinct swimming behaviors including backward swimming. These approaches provide new opportunities for the optogenetic dissection of neuronal circuit structure and function.

20.
Nat Neurosci ; 10(12): 1569-77, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18026099

RESUMO

Functional maturation of GABAergic innervation in the developing visual cortex is regulated by neural activity and sensory inputs and in turn influences the critical period of ocular dominance plasticity. Here we show that polysialic acid (PSA), presented by the neural cell adhesion molecule, has a role in the maturation of GABAergic innervation and ocular dominance plasticity. Concentrations of PSA significantly decline shortly after eye opening in the adolescent mouse visual cortex; this decline is hindered by visual deprivation. The developmental and activity-dependent regulation of PSA expression is inversely correlated with the maturation of GABAergic innervation. Premature removal of PSA in visual cortex results in precocious maturation of perisomatic innervation by basket interneurons, enhanced inhibitory synaptic transmission, and earlier onset of ocular dominance plasticity. The developmental and activity-dependent decline of PSA expression therefore regulates the timing of the maturation of GABAergic inhibition and the onset of ocular dominance plasticity.


Assuntos
Período Crítico Psicológico , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Inibição Neural/fisiologia , Plasticidade Neuronal/fisiologia , Ácidos Siálicos/metabolismo , Fatores Etários , Análise de Variância , Anestésicos Locais/farmacologia , Animais , Animais Recém-Nascidos , Potenciais Evocados Visuais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Glutamato Descarboxilase/metabolismo , Glicosídeo Hidrolases/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Camundongos , Moléculas de Adesão de Célula Nervosa/metabolismo , Estimulação Luminosa/métodos , Privação Sensorial/fisiologia , Ácidos Siálicos/antagonistas & inibidores , Tetrodotoxina/farmacologia , Córtex Visual/crescimento & desenvolvimento , Vias Visuais/fisiologia , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...