Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(4)2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36810252

RESUMO

Angelman syndrome (AS) is a severe neurodevelopmental disorder (NDD) caused by loss of functional ubiquitin protein ligase E3A (UBE3A). Previous studies showed that UBE3A plays an important role in the first postnatal weeks of mouse brain development, but its precise role is unknown. Since impaired striatal maturation has been implicated in several mouse models for NDDs, we studied the importance of UBE3A in striatal maturation. We used inducible Ube3a mouse models to investigate the maturation of medium spiny neurons (MSNs) from dorsomedial striatum. MSNs of mutant mice matured properly till postnatal day 15 (P15) but remained hyperexcitable with fewer excitatory synaptic events at later ages, indicative of stalled striatal maturation in Ube3a mice. Reinstatement of UBE3A expression at P21 fully restored MSN excitability but only partially restored synaptic transmission and the operant conditioning behavioral phenotype. Gene reinstatement at P70 failed to rescue both electrophysiological and behavioral phenotypes. In contrast, deletion of Ube3a after normal brain development did not result in these electrophysiological and behavioral phenotypes. This study emphasizes the role of UBE3A in striatal maturation and the importance of early postnatal reinstatement of UBE3A expression to obtain a full rescue of behavioral phenotypes associated with striatal function in AS.


Assuntos
Síndrome de Angelman , Encéfalo , Ubiquitina-Proteína Ligases , Animais , Camundongos , Síndrome de Angelman/genética , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
2.
JCI Insight ; 6(23)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34877936

RESUMO

Tuberous sclerosis complex (TSC), caused by heterozygous mutations in TSC1 or TSC2, frequently results in intractable epilepsy. Here, we made use of an inducible Tsc1-knockout mouse model, allowing us to study electrophysiological and molecular changes of Tsc1-induced epileptogenesis over time. We recorded from pyramidal neurons in the hippocampus and somatosensory cortex (L2/L3) and combined this with an analysis of transcriptome changes during epileptogenesis. Deletion of Tsc1 resulted in hippocampus-specific changes in excitability and adaptation, which emerged before seizure onset and progressed over time. All phenotypes were rescued after early treatment with rapamycin, an mTOR inhibitor. Later in epileptogenesis, we observed a hippocampal increase of excitation-to-inhibition ratio. These cellular changes were accompanied by dramatic transcriptional changes, especially after seizure onset. Most of these changes were rescued upon rapamycin treatment. Of the genes encoding ion channels or belonging to the Gene Ontology term action potential, 27 were differentially expressed just before seizure onset, suggesting a potential driving role in epileptogenesis. Our data highlight the complex changes driving epileptogenesis in TSC, including the changed expression of multiple ion channels. Our study emphasizes inhibition of the TSC/mTOR signaling pathway as a promising therapeutic approach to target epilepsy in patients with TSC.


Assuntos
Epilepsia/genética , Esclerose Tuberosa/genética , Animais , Modelos Animais de Doenças , Humanos , Canais Iônicos , Masculino , Camundongos , Camundongos Knockout , Esclerose Tuberosa/patologia
3.
Neuroscience ; 445: 172-189, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32088294

RESUMO

The UBE3A gene is part of the chromosome 15q11-q13 region that is frequently deleted or duplicated, leading to several neurodevelopmental disorders (NDD). Angelman syndrome (AS) is caused by the absence of functional maternally derived UBE3A protein, while the paternal UBE3A gene is present but silenced specifically in neurons. Patients with AS present with severe neurodevelopmental delay, with pronounced motor deficits, absence of speech, intellectual disability, epilepsy, and sleep problems. The pathophysiology of AS is still unclear and a treatment is lacking. Animal models of AS recapitulate the genotypic and phenotypic features observed in AS patients, and have been invaluable for understanding the disease process as well as identifying apropriate drug targets. Using these AS mouse models we have learned that loss of UBE3A probably affects many areas of the brain, leading to increased neuronal excitability and a loss of synaptic spines, along with changes in a number of distinct behaviours. Inducible AS mouse models have helped to identify the critical treatment windows for the behavioral and physiological phenotypes. Additionally, AS mouse models indicate an important role for the predominantly nuclear UBE3A isoform in generating the characteristic AS pathology. Last, but not least, the AS mice have been crucial in guiding Ube3a gene reactivation treatments, which present a very promising therapy to treat AS.


Assuntos
Síndrome de Angelman , Síndrome de Angelman/genética , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Neurônios , Fenótipo , Ubiquitina-Proteína Ligases/genética
4.
Nat Neurosci ; 22(8): 1235-1247, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31235931

RESUMO

Mutations affecting the gene encoding the ubiquitin ligase UBE3A cause Angelman syndrome. Although most studies focus on the synaptic function of UBE3A, we show that UBE3A is highly enriched in the nucleus of mouse and human neurons. We found that the two major isoforms of UBE3A exhibit highly distinct nuclear versus cytoplasmic subcellular localization. Both isoforms undergo nuclear import through direct binding to PSMD4 (also known as S5A or RPN10), but the amino terminus of the cytoplasmic isoform prevents nuclear retention. Mice lacking the nuclear UBE3A isoform recapitulate the behavioral and electrophysiological phenotypes of Ube3am-/p+ mice, whereas mice harboring a targeted deletion of the cytosolic isoform are unaffected. Finally, we identified Angelman syndrome-associated UBE3A missense mutations that interfere with either nuclear targeting or nuclear retention of UBE3A. Taken together, our findings elucidate the mechanisms underlying the subcellular localization of UBE3A, and indicate that the nuclear UBE3A isoform is the most critical for the pathophysiology of Angelman syndrome.


Assuntos
Síndrome de Angelman/genética , Síndrome de Angelman/psicologia , Comportamento Animal , Ubiquitina-Proteína Ligases/genética , Animais , Proteínas de Transporte/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/genética , Citosol/enzimologia , Fenômenos Eletrofisiológicos/genética , Feminino , Humanos , Isoenzimas/genética , Masculino , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto/genética , Comportamento de Nidação , Neurônios/enzimologia , Desempenho Psicomotor , Proteínas de Ligação a RNA , Natação/psicologia , Dedos de Zinco
5.
J Neurosci ; 38(37): 8011-8030, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30082419

RESUMO

E3 ubiquitin ligase (UBE3A) levels in the brain need to be tightly regulated, as loss of functional UBE3A protein is responsible for the severe neurodevelopmental disorder Angelman syndrome (AS), whereas increased activity of UBE3A is associated with nonsyndromic autism. Given the role of mPFC in neurodevelopmental disorders including autism, we aimed to identify the functional changes resulting from loss of UBE3A in infralimbic and prelimbic mPFC areas in a mouse model of AS. Whole-cell recordings from layer 5 mPFC pyramidal neurons obtained in brain slices from adult mice of both sexes revealed that loss of UBE3A results in a strong decrease of spontaneous inhibitory transmission and increase of spontaneous excitatory transmission potentially leading to a marked excitation/inhibition imbalance. Additionally, we found that loss of UBE3A led to decreased excitability and increased threshold for action potential of layer 5 fast spiking interneurons without significantly affecting the excitability of pyramidal neurons. Because we previously showed that AS mouse behavioral phenotypes are reversible upon Ube3a gene reactivation during a restricted period of early postnatal development, we investigated whether Ube3a gene reactivation in a fully mature brain could reverse any of the identified physiological deficits. In contrast to our previously reported behavioral findings, restoring UBE3A levels in adult animals fully rescued all the identified physiological deficits of mPFC neurons. Moreover, the kinetics of reversing these synaptic deficits closely followed the reinstatement of UBE3A protein level. Together, these findings show a striking dissociation between the rescue of behavioral and physiological deficits.SIGNIFICANCE STATEMENT Here we describe significant physiological deficits in the mPFC of an Angelman syndrome mouse model. We found a marked change in excitatory/inhibitory balance, as well as decreased excitability of fast spiking interneurons. A promising treatment strategy for Angelman syndrome is aimed at restoring UBE3A expression by activating the paternal UBE3A gene. Here we find that the physiological changes in the mPFC are fully reversible upon gene reactivation, even when the brain is fully mature. This indicates that there is no critical developmental window for reversing the identified physiological deficits in mPFC.


Assuntos
Síndrome de Angelman/fisiopatologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiopatologia , Ubiquitina-Proteína Ligases/genética , Potenciais de Ação/fisiologia , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Técnicas de Patch-Clamp , Ubiquitina-Proteína Ligases/metabolismo
6.
Nat Commun ; 7: 10682, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26931375

RESUMO

Trafficking and biophysical properties of AMPA receptors (AMPARs) in the brain depend on interactions with associated proteins. We identify Shisa6, a single transmembrane protein, as a stable and directly interacting bona fide AMPAR auxiliary subunit. Shisa6 is enriched at hippocampal postsynaptic membranes and co-localizes with AMPARs. The Shisa6 C-terminus harbours a PDZ domain ligand that binds to PSD-95, constraining mobility of AMPARs in the plasma membrane and confining them to postsynaptic densities. Shisa6 expressed in HEK293 cells alters GluA1- and GluA2-mediated currents by prolonging decay times and decreasing the extent of AMPAR desensitization, while slowing the rate of recovery from desensitization. Using gene deletion, we show that Shisa6 increases rise and decay times of hippocampal CA1 miniature excitatory postsynaptic currents (mEPSCs). Shisa6-containing AMPARs show prominent sustained currents, indicating protection from full desensitization. Accordingly, Shisa6 prevents synaptically trapped AMPARs from depression at high-frequency synaptic transmission.


Assuntos
Hipocampo/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/fisiologia , Receptores de AMPA/metabolismo , Animais , Células Cultivadas , Fenômenos Eletrofisiológicos , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Hipocampo/citologia , Humanos , Proteínas de Membrana/genética , Camundongos , Neurônios/citologia , Ratos , Receptores de AMPA/genética , Sinapses , Técnicas do Sistema de Duplo-Híbrido
7.
J Biol Chem ; 290(25): 15635-15645, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25911096

RESUMO

The inbred strains C57BL/6J and DBA/2J (DBA) display striking differences in a number of behavioral tasks depending on hippocampal function, such as contextual memory. Historically, this has been explained through differences in postsynaptic protein expression underlying synaptic transmission and plasticity. We measured the synaptic hippocampal protein content (iTRAQ (Isobaric Tags for Relative and Absolute Quantitation) and mass spectrometry), CA1 synapse ultrastructural morphology, and synaptic functioning in adult C57BL/6J and DBA mice. DBA mice showed a prominent decrease in the Ras-GAP calcium-sensing protein RASAL1. Furthermore, expression of several presynaptic markers involved in exocytosis, such as syntaxin (Stx1b), Ras-related proteins (Rab3a/c), and rabphilin (Rph3a), was reduced. Ultrastructural analysis of CA1 hippocampal synapses showed a significantly lower number of synaptic vesicles and presynaptic cluster size in DBA mice, without changes in postsynaptic density or active zone. In line with this compromised presynaptic morphological and molecular phenotype in DBA mice, we found significantly lower paired-pulse facilitation and enhanced short term depression of glutamatergic synapses, indicating a difference in transmitter release and/or refilling mechanisms. Taken together, our data suggest that in addition to strain-specific postsynaptic differences, the change in dynamic properties of presynaptic transmitter release may underlie compromised synaptic processing related to cognitive functioning in DBA mice.


Assuntos
Cognição/fisiologia , Hipocampo , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Densidade Pós-Sináptica , Proteoma/metabolismo , Animais , Hipocampo/fisiologia , Hipocampo/ultraestrutura , Camundongos , Camundongos Endogâmicos DBA , Proteoma/fisiologia , Proteoma/ultraestrutura , Proteômica , Especificidade da Espécie
8.
J Neurophysiol ; 113(6): 1850-61, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25540225

RESUMO

In rodent cortex GABAA receptor (GABAAR)-mediated synapses are a significant source of input onto GABA neurons, and the properties of these inputs vary among GABA neuron subtypes that differ in molecular markers and firing patterns. Some features of cortical interneurons are different between rodents and primates, but it is not known whether inhibition of GABA neurons is prominent in the primate cortex and, if so, whether these inputs show heterogeneity across GABA neuron subtypes. We thus studied GABAAR-mediated miniature synaptic events in GABAergic interneurons in layer 3 of monkey dorsolateral prefrontal cortex (DLPFC). Interneurons were identified on the basis of their firing pattern as fast spiking (FS), regular spiking (RS), burst spiking (BS), or irregular spiking (IS). Miniature synaptic events were common in all of the recorded interneurons, and the frequency of these events was highest in FS neurons. The amplitude and kinetics of miniature inhibitory postsynaptic potentials (mIPSPs) also differed between DLPFC interneuron subtypes in a manner correlated with their input resistance and membrane time constant. FS neurons had the fastest mIPSP decay times and the strongest effects of the GABAAR modulator zolpidem, suggesting that the distinctive properties of inhibitory synaptic inputs onto FS cells are in part conferred by GABAARs containing α1 subunits. Moreover, mIPSCs differed between FS and RS interneurons in a manner consistent with the mIPSP findings. These results show that in the monkey DLPFC GABAAR-mediated synaptic inputs are prominent in layer 3 interneurons and may differentially regulate the activity of different interneuron subtypes.


Assuntos
Potenciais de Ação , Neurônios GABAérgicos/metabolismo , Córtex Pré-Frontal/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Feminino , Agonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/fisiologia , Potenciais Pós-Sinápticos Inibidores , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Interneurônios/fisiologia , Macaca mulatta , Masculino , Potenciais Pós-Sinápticos em Miniatura , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/fisiologia , Piridinas/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Zolpidem
9.
Cereb Cortex ; 25(11): 4076-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24904071

RESUMO

Development of inhibition onto pyramidal cells may be crucial for the emergence of cortical network activity, including gamma oscillations. In primate dorsolateral prefrontal cortex (DLPFC), inhibitory synaptogenesis starts in utero and inhibitory synapse density reaches adult levels before birth. However, in DLPFC, the expression levels of γ-aminobutyric acid (GABA) synapse-related gene products changes markedly during development until young adult age, suggesting a highly protracted maturation of GABA synapse function. Therefore, we examined the development of GABA synapses by recording GABAAR-mediated inhibitory postsynaptic currents (GABAAR-IPSCs) from pyramidal cells in the DLPFC of neonatal, prepubertal, peripubertal, and adult macaque monkeys. We found that the decay of GABAAR-IPSCs, possibly including those from parvalbumin-positive GABA neurons, shortened by prepubertal age, while their amplitude increased until the peripubertal period. Interestingly, both GABAAR-mediated quantal response size, estimated by miniature GABAAR-IPSCs, and the density of GABAAR synaptic appositions, measured with immunofluorescence microscopy, were stable with age. Simulations in a computational model network with constant GABA synapse density showed that the developmental changes in GABAAR-IPSC properties had a significant impact on oscillatory activity and predicted that, whereas DLPFC circuits can generate gamma frequency oscillations by prepubertal age, mature levels of gamma band power are attained at late stages of development.


Assuntos
Potenciais Pós-Sinápticos Inibidores/fisiologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Sinapses/fisiologia , Ácido gama-Aminobutírico/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Bloqueadores dos Canais de Cálcio/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Antagonistas GABAérgicos/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Lisina/análogos & derivados , Lisina/metabolismo , Macaca mulatta , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Piridazinas/farmacologia , Sinapses/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , ômega-Agatoxina IVA/farmacologia
10.
J Neurosci ; 34(49): 16234-46, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25471564

RESUMO

The basal forebrain cholinergic innervation of the medial prefrontal cortex (mPFC) is crucial for cognitive performance. However, little is known about the organization of connectivity between the basal forebrain and the mPFC in the mouse. Using focal virus injections inducing Cre-dependent enhanced yellow fluorescent protein expression in ChAT-IRES-Cre mice, we tested the hypothesis that there is a topographic mapping between the basal forebrain cholinergic neurons and their axonal projections to the mPFC. We found that ascending cholinergic fibers to the mPFC follow four pathways and that cholinergic neurons take these routes depending on their location in the basal forebrain. In addition, a general mapping pattern was observed in which the position of cholinergic neurons measured along a rostral to caudal extent in the basal forebrain correlated with a ventral to dorsal and a rostral to caudal shift of cholinergic fiber distribution in mPFC. Finally, we found that neurons in the rostral and caudal parts of the basal forebrain differentially innervate the superficial and deep layers of the ventral regions of the mPFC. Thus, a frontocaudal organization of the cholinergic system exists in which distinct mPFC areas and cortical layers are targeted depending on the location of the cholinergic neuron in the basal forebrain.


Assuntos
Prosencéfalo Basal/anatomia & histologia , Prosencéfalo Basal/citologia , Mapeamento Encefálico , Neurônios Colinérgicos , Córtex Pré-Frontal/anatomia & histologia , Córtex Pré-Frontal/citologia , Animais , Camundongos , Vias Neurais/anatomia & histologia , Vias Neurais/citologia , Técnicas de Rastreamento Neuroanatômico
11.
J Neurosci ; 33(46): 18225-33, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24227731

RESUMO

In addicts, associative memories related to the rewarding effects of drugs of abuse can evoke powerful craving and drug seeking urges, but effective treatment to suppress these memories is not available. Detailed insight into the neural circuitry that mediates expression of drug-associated memory is therefore of crucial importance. Substantial evidence from rodent models of addictive behavior points to the involvement of the ventromedial prefrontal cortex (vmPFC) in conditioned drug seeking, but specific knowledge of the temporal role of vmPFC pyramidal cells is lacking. To this end, we used an optogenetics approach to probe the involvement of vmPFC pyramidal cells in expression of a recent and remote conditioned cocaine memory. In mice, we expressed Channelrhodopsin-2 (ChR2) or Halorhodopsin (eNpHR3.0) in pyramidal cells of the vmPFC and studied the effect of activation or inhibition of these cells during expression of a cocaine-contextual memory on days 1-2 (recent) and ∼3 weeks (remote) after conditioning. Whereas optical activation of pyramidal cells facilitated extinction of remote memory, without affecting recent memory, inhibition of pyramidal cells acutely impaired recall of recent cocaine memory, without affecting recall of remote memory. In addition, we found that silencing pyramidal cells blocked extinction learning at the remote memory time-point. We provide causal evidence of a critical time-dependent switch in the contribution of vmPFC pyramidal cells to recall and extinction of cocaine-associated memory, indicating that the circuitry that controls expression of cocaine memories reorganizes over time.


Assuntos
Cocaína/farmacologia , Extinção Psicológica/fisiologia , Rememoração Mental/fisiologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Animais , Extinção Psicológica/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Distribuição Aleatória , Fatores de Tempo
12.
Rev Neurosci ; 23(1): 97-109, 2012 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-22718616

RESUMO

Cognitive impairment, a core feature of schizophrenia, has been suggested to arise from a disturbance of gamma oscillations that is due to decreased neurotransmission from the parvalbumin (PV) subtype of interneurons. Indeed, PV interneurons have uniquely fast membrane and synaptic properties that are crucially important for network functions such as feedforward inhibition or gamma oscillations. The causes leading to impairment of PV neurotransmission in schizophrenia are still under investigation. Interestingly, NMDA receptors (NMDARs) antagonism results in schizophrenia-like symptoms in healthy adults. Additionally, systemic NMDAR antagonist administration increases prefrontal cortex pyramidal cell firing, apparently by producing disinhibition, and repeated exposure to NMDA antagonists leads to changes in the GABAergic markers that mimic the impairments found in schizophrenia. Based on these findings, PV neuron deficits in schizophrenia have been proposed to be secondary to (NMDAR) hypofunction at glutamatergic synapses onto these cells. However, NMDARs generate long-lasting postsynaptic currents that result in prolonged depolarization of the postsynaptic cells, a property inconsistent with the role of PV cells in network dynamics. Here, we review evidence leading to the conclusion that cortical disinhibition and GABAergic impairment produced by NMDAR antagonists are unlikely to be mediated via NMDARs at glutamatergic synapses onto mature cortical PV neurons.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Ácido Glutâmico/metabolismo , Interneurônios/metabolismo , Parvalbuminas/metabolismo , Doença de Alzheimer/metabolismo , Animais , Humanos , Vias Neurais
13.
Nat Neurosci ; 14(10): 1302-8, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909089

RESUMO

Upon retrieval, fear memories are rendered labile and prone to modification, necessitating a restabilization process of reconsolidation to persist further. This process is also crucial for modulating both strength and content of an existing memory and forms a promising therapeutic target for fear-related disorders. However, the molecular and cellular mechanism of adaptive reconsolidation still remains obscure. Here we show that retrieval of fear memory induces a biphasic temporal change in GluA2-containing AMPA-type glutamate receptor (AMPAR) membrane expression and synaptic strength in the mouse dorsal hippocampus. Blockade of retrieval-induced, regulated, GluA2-dependent endocytosis enhanced subsequent expression of fear. In addition, this blockade prevented the loss of fear response after reconsolidation-update of fear memory content in the long-term. Thus, endocytosis of GluA2-containing AMPARs allows plastic changes at the synaptic level that exerts an inhibitory constraint on memory strengthening and underlies the loss of fear response by reinterpretation of memory content during adaptive reconsolidation.


Assuntos
Condicionamento Clássico/fisiologia , Endocitose/fisiologia , Medo/fisiologia , Rememoração Mental/fisiologia , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Análise de Variância , Animais , Anisomicina/farmacologia , Comportamento Animal , Biotinilação , Condicionamento Clássico/efeitos dos fármacos , Eletrochoque/efeitos adversos , Endocitose/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Técnicas In Vitro , Masculino , Rememoração Mental/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico/efeitos dos fármacos , Receptores de AMPA/química , Sinapses/efeitos dos fármacos , Fatores de Tempo
14.
J Biol Chem ; 286(29): 25495-504, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21596744

RESUMO

Fragile X syndrome (FXS), the most common form of hereditary mental retardation, is caused by a loss-of-function mutation of the Fmr1 gene, which encodes fragile X mental retardation protein (FMRP). FMRP affects dendritic protein synthesis, thereby causing synaptic abnormalities. Here, we used a quantitative proteomics approach in an FXS mouse model to reveal changes in levels of hippocampal synapse proteins. Sixteen independent pools of Fmr1 knock-out mice and wild type mice were analyzed using two sets of 8-plex iTRAQ experiments. Of 205 proteins quantified with at least three distinct peptides in both iTRAQ series, the abundance of 23 proteins differed between Fmr1 knock-out and wild type synapses with a false discovery rate (q-value) <5%. Significant differences were confirmed by quantitative immunoblotting. A group of proteins that are known to be involved in cell differentiation and neurite outgrowth was regulated; they included Basp1 and Gap43, known PKC substrates, and Cend1. Basp1 and Gap43 are predominantly expressed in growth cones and presynaptic terminals. In line with this, ultrastructural analysis in developing hippocampal FXS synapses revealed smaller active zones with corresponding postsynaptic densities and smaller pools of clustered vesicles, indicative of immature presynaptic maturation. A second group of proteins involved in synaptic vesicle release was up-regulated in the FXS mouse model. In accordance, paired-pulse and short-term facilitation were significantly affected in these hippocampal synapses. Together, the altered regulation of presynaptically expressed proteins, immature synaptic ultrastructure, and compromised short-term plasticity points to presynaptic changes underlying glutamatergic transmission in FXS at this stage of development.


Assuntos
Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Fenótipo , Proteômica , Sinapses/metabolismo , Actinas/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Região CA1 Hipocampal/ultraestrutura , Diferenciação Celular , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Neuritos/metabolismo , Plasticidade Neuronal/fisiologia , Pseudópodes/metabolismo , Sinapses/patologia , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/patologia , Espectrometria de Massas em Tandem
15.
J Neurosci ; 31(1): 142-56, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21209199

RESUMO

Schizophrenia may involve hypofunction of NMDA receptor (NMDAR)-mediated signaling, and alterations in parvalbumin-positive fast-spiking (FS) GABA neurons that may cause abnormal gamma oscillations. It was recently hypothesized that prefrontal cortex (PFC) FS neuron activity is highly dependent on NMDAR activation and that, consequently, FS neuron dysfunction in schizophrenia is secondary to NMDAR hypofunction. However, NMDARs are abundant in synapses onto PFC pyramidal neurons; thus, a key question is whether FS neuron or pyramidal cell activation is more dependent on NMDARs. We examined the AMPAR and NMDAR contribution to synaptic activation of FS neurons and pyramidal cells in the PFC of adult mice. In FS neurons, EPSCs had fast decay and weak NMDAR contribution, whereas in pyramidal cells, EPSCs were significantly prolonged by NMDAR-mediated currents. Moreover, the AMPAR/NMDAR EPSC ratio was higher in FS cells. NMDAR antagonists decreased EPSPs and EPSP-spike coupling more strongly in pyramidal cells than in FS neurons, showing that FS neuron activation is less NMDAR dependent than pyramidal cell excitation. The precise EPSP-spike coupling produced by fast-decaying EPSCs in FS cells may be important for network mechanisms of gamma oscillations based on feedback inhibition. To test this possibility, we used simulations in a computational network of reciprocally connected FS neurons and pyramidal cells and found that brief AMPAR-mediated FS neuron activation is crucial to synchronize, via feedback inhibition, pyramidal cells in the gamma frequency band. Our results raise interesting questions about the mechanisms that might link NMDAR hypofunction to alterations of FS neurons in schizophrenia.


Assuntos
Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Receptores de Glutamato/metabolismo , Sinapses/metabolismo , Potenciais de Ação/fisiologia , Animais , Biofísica/métodos , Simulação por Computador , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Neurônios/classificação , Técnicas de Patch-Clamp/métodos , Probabilidade , Piridazinas/farmacologia , Receptores de Glutamato/genética , Estatísticas não Paramétricas
16.
J Neurophysiol ; 101(2): 533-47, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19073797

RESUMO

The plasma membrane GABA transporter GAT1 is thought to mediate uptake of synaptically released GABA. In the primate dorsolateral prefrontal cortex (DLPFC), GAT1 expression changes significantly during development and in schizophrenia. The consequences of such changes, however, are not well understood because GAT1's role has not been investigated in primate neocortical circuits. We thus studied the effects of the GAT1 blocker 1,2,5,6-tetrahydro-1-[2-[[(diphenylmethylene)amino]oxy]ethyl]-3-pyridinecarboxylic acid hydrochloride (NO711) on GABA transmission onto pyramidal neurons of monkey DLPFC. As in rat cortex, in monkey DLPFC NO711 did not substantially alter miniature GABA transmission, suggesting that GAT1 does not regulate single-synapse transmission. In rat cortical circuits, between-synapse GABA spillover produced by NO711 clearly prolongs the inhibitory postsynaptic currents, but whether NO711 also prolongs the inhibitory postsynaptic potentials (IPSPs) is unclear. Moreover, whether spillover differentially affects perisomatic versus dendritic inputs has not been examined. Here we found that NO711 prolonged the GABAA receptor-mediated IPSPs (GABAAR-IPSPs) evoked by stimulating perisomatic synapses. Dendritic, but not perisomatic, synapse stimulation often elicited a postsynaptic GABAB receptor-mediated IPSP that was enhanced by NO711. Blocking GABAB receptors revealed that NO711 prolonged the GABAAR-IPSPs evoked by stimulation of dendrite-targeting inputs. We conclude that a major functional role for GAT1 in primate cortical circuits is to prevent the effects of GABA spillover when multiple synapses are simultaneously active. Furthermore, we report that, at least in monkey DLPFC, GAT1 similarly restricts GABA spillover onto perisomatic or dendritic inputs, critically controlling the spatiotemporal specificity of inhibitory inputs onto proximal or distal compartments of the pyramidal cell membrane.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/citologia , Sinapses/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Estimulação Elétrica , Feminino , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Inibidores da Captação de GABA , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Macaca fascicularis , Macaca mulatta , Masculino , Modelos Neurológicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ácidos Nipecóticos/farmacologia , Compostos Organofosforados/farmacologia , Oximas/farmacologia , Técnicas de Patch-Clamp/métodos , Piridazinas/farmacologia , Sinapses/efeitos dos fármacos
17.
J Physiol ; 581(Pt 3): 981-1000, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17395630

RESUMO

Dopamine (DA) effects on prefrontal cortex (PFC) neurons are essential for the cognitive functions mediated by this cortical area. However, the cellular mechanisms of DA neuromodulation in neocortex are not well understood. We characterized the effects of D1-type DA receptor (D1R) activation on the amplification (increase in duration and area) of excitatory postsynaptic potentials (EPSPs) at depolarized potentials, in layer 5 pyramidal neurons from rat PFC. Simulated EPSPs (sEPSPs) were elicited by current injection, to determine the effects of D1R activation independent of modulation of transmitter release or glutamate receptor currents. Application of the D1R agonist SKF81297 attenuated sEPSP amplification at depolarized potentials in a concentration-dependent manner. The SKF81297 effects were inhibited by the D1R antagonist SCH23390. The voltage-gated Na+ channel blocker tetrodotoxin (TTX) abolished the effects of SKF81297 on sEPSP amplification, suggesting that Na+ currents are necessary for the D1R effect. Furthermore, blockade of 4-AP- and TEA-sensitive K+ channels in the presence of TTX significantly increased EPSP amplification, arguing against the possibility that SKF81297 up-regulates currents that attenuate sEPSP amplification. SKF81297 application attenuated the subthreshold response to injection of depolarizing current ramps, in a manner consistent with a decrease in the persistent Na+ current. In addition, D1R activation decreased the effectiveness of temporal EPSP summation during 20 Hz sEPSP trains, selectively at depolarized membrane potentials. Therefore, the effects of D1R activation on Na+ channel-dependent EPSP amplification may regulate the impact of coincidence detection versus temporal integration mechanisms in PFC pyramidal neurons.


Assuntos
Dopamina/metabolismo , Potenciais Pós-Sinápticos Excitadores , Plasticidade Neuronal , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Receptores de Dopamina D1/metabolismo , Canais de Sódio/metabolismo , Sinapses/metabolismo , Animais , Benzazepinas/farmacologia , Cognição , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Técnicas In Vitro , Masculino , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Dopamina D1/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Tetrodotoxina/farmacologia , Fatores de Tempo
18.
J Comp Neurol ; 490(3): 220-38, 2005 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16082676

RESUMO

The mediodorsal nucleus of the thalamus (MD) represents the main subcortical structure that projects to the prefrontal cortex (PFC) and it regulates key aspects of the cognitive functions of this region. Within the PFC, GABA local circuit neurons shape the activity patterns and hence the "memory fields" of pyramidal cells. Although the connections between the MD and PFC are well established, the ultrastructural relationships between projecting fibers from the MD and different subclasses of GABA cells in the PFC are not known. In order to address this issue in the rat, we examined MD axons labeled by tract-tracing in combination with immunogold-silver to identify different calcium-binding proteins localized within separate populations of interneurons. Electron micrographic examination of PFC sections from these animals revealed that MD terminals made primarily asymmetric synapses onto dendritic spines and less commonly onto dendritic shafts. Most of the dendrites receiving MD synaptic input were immunoreactive for parvalbumin (ParV), whereas MD synapses onto dendrites labeled for calretinin or calbindin were less frequent. We also observed that some MD terminals were themselves immunoreactive for calcium-binding proteins, again more commonly for ParV. These results suggest that the MD exerts a dual influence on PFC pyramidal cells: direct inputs onto spines and an indirect influence mediated via synapses onto each subclass of interneurons. The apparent preferential input to ParV cells endows MD afferents with a strong indirect inhibitory influence on pyramidal neuron activity by virtue of ParV cell synapses onto soma, proximal dendrites, and axon initial segments.


Assuntos
Vias Aferentes/anatomia & histologia , Interneurônios/citologia , Córtex Pré-Frontal/citologia , Sinapses/metabolismo , Tálamo/citologia , Vias Aferentes/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Imuno-Histoquímica/métodos , Interneurônios/metabolismo , Interneurônios/ultraestrutura , Masculino , Microscopia Imunoeletrônica/métodos , Modelos Biológicos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Sinapses/ultraestrutura , Tálamo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...