Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 286(37): 32713-22, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21771791

RESUMO

Aberrant microglial activation has been proposed to contribute to the cognitive decline in Alzheimer disease (AD), but the underlying molecular mechanisms remain enigmatic. Fractalkine signaling, a pathway mediating the communication between microglia and neurons, is deficient in AD brains and down-regulated by amyloid-ß. Although fractalkine receptor (CX3CR1) on microglia was found to regulate plaque load, no functional effects have been reported. Our study demonstrates that CX3CR1 deficiency worsens the AD-related neuronal and behavioral deficits. The effects were associated with cytokine production but not with plaque deposition. Ablation of CX3CR1 in mice overexpressing human amyloid precursor protein enhanced Tau pathology and exacerbated the depletion of calbindin in the dentate gyrus. The levels of calbindin in the dentate gyrus correlated negatively with those of tumor necrosis factor α and interleukin 6, suggesting neurotoxic effects of inflammatory factors. Functionally, removing CX3CR1 in human amyloid precursor protein mice worsened the memory retention in passive avoidance and novel object recognition tests, and their memory loss in the novel object recognition test is associated with high levels of interleukin 6. Our findings identify CX3CR1 as a key microglial pathway in protecting against AD-related cognitive deficits that are associated with aberrant microglial activation and elevated inflammatory cytokines.


Assuntos
Doença de Alzheimer/metabolismo , Transtornos Cognitivos/metabolismo , Microglia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Quimiocinas/metabolismo , Transdução de Sinais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Receptor 1 de Quimiocina CX3C , Calbindinas , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Citocinas/genética , Citocinas/metabolismo , Giro Denteado/metabolismo , Giro Denteado/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Microglia/patologia , Proteínas do Tecido Nervoso/genética , Receptores de Quimiocinas/genética , Proteína G de Ligação ao Cálcio S100/genética , Proteína G de Ligação ao Cálcio S100/metabolismo
2.
J Neurosci ; 31(2): 700-11, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228179

RESUMO

Alzheimer's disease (AD), the most common neurodegenerative disorder, is a growing public health problem and still lacks effective treatments. Recent evidence suggests that microtubule-associated protein tau may mediate amyloid-ß peptide (Aß) toxicity by modulating the tyrosine kinase Fyn. We showed previously that tau reduction prevents, and Fyn overexpression exacerbates, cognitive deficits in human amyloid precursor protein (hAPP) transgenic mice overexpressing Aß. However, the mechanisms by which Aß, tau, and Fyn cooperate in AD-related pathogenesis remain to be fully elucidated. Here we examined the synaptic and network effects of this pathogenic triad. Tau reduction prevented cognitive decline induced by synergistic effects of Aß and Fyn. Tau reduction also prevented synaptic transmission and plasticity deficits in hAPP mice. Using electroencephalography to examine network effects, we found that tau reduction prevented spontaneous epileptiform activity in multiple lines of hAPP mice. Tau reduction also reduced the severity of spontaneous and chemically induced seizures in mice overexpressing both Aß and Fyn. To better understand these protective effects, we recorded whole-cell currents in acute hippocampal slices from hAPP mice with and without tau. hAPP mice with tau had increased spontaneous and evoked excitatory currents, reduced inhibitory currents, and NMDA receptor dysfunction. Tau reduction increased inhibitory currents and normalized excitation/inhibition balance and NMDA receptor-mediated currents in hAPP mice. Our results indicate that Aß, tau, and Fyn jointly impair synaptic and network function and suggest that disrupting the copathogenic relationship between these factors could be of therapeutic benefit.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/fisiologia , Transtornos Cognitivos/fisiopatologia , Rede Nervosa/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Sinapses/fisiologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/mortalidade , Animais , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/psicologia , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Hipocampo/fisiopatologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Mutantes , Plasticidade Neuronal , Convulsões/metabolismo , Convulsões/fisiopatologia , Especificidade da Espécie , Transmissão Sináptica , Proteínas tau/genética
3.
Nature ; 469(7328): 47-52, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21113149

RESUMO

Amyloid-ß oligomers may cause cognitive deficits in Alzheimer's disease by impairing neuronal NMDA-type glutamate receptors, whose function is regulated by the receptor tyrosine kinase EphB2. Here we show that amyloid-ß oligomers bind to the fibronectin repeats domain of EphB2 and trigger EphB2 degradation in the proteasome. To determine the pathogenic importance of EphB2 depletions in Alzheimer's disease and related models, we used lentiviral constructs to reduce or increase neuronal expression of EphB2 in memory centres of the mouse brain. In nontransgenic mice, knockdown of EphB2 mediated by short hairpin RNA reduced NMDA receptor currents and impaired long-term potentiation in the dentate gyrus, which are important for memory formation. Increasing EphB2 expression in the dentate gyrus of human amyloid precursor protein transgenic mice reversed deficits in NMDA receptor-dependent long-term potentiation and memory impairments. Thus, depletion of EphB2 is critical in amyloid-ß-induced neuronal dysfunction. Increasing EphB2 levels or function could be beneficial in Alzheimer's disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Cognição/fisiologia , Receptor EphB2/deficiência , Receptor EphB2/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Giro Denteado/metabolismo , Modelos Animais de Doenças , Humanos , Potenciação de Longa Duração , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Receptor EphB2/química , Receptor EphB2/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo
4.
Neuron ; 68(3): 428-41, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21040845

RESUMO

The entorhinal cortex (EC) is one of the earliest affected, most vulnerable brain regions in Alzheimer's disease (AD), which is associated with amyloid-ß (Aß) accumulation in many brain areas. Selective overexpression of mutant amyloid precursor protein (APP) predominantly in layer II/III neurons of the EC caused cognitive and behavioral abnormalities characteristic of mouse models with widespread neuronal APP overexpression, including hyperactivity, disinhibition, and spatial learning and memory deficits. APP/Aß overexpression in the EC elicited abnormalities in synaptic functions and activity-related molecules in the dentate gyrus and CA1 and epileptiform activity in parietal cortex. Soluble Aß was observed in the dentate gyrus, and Aß deposits in the hippocampus were localized to perforant pathway terminal fields. Thus, APP/Aß expression in EC neurons causes transsynaptic deficits that could initiate the cortical-hippocampal network dysfunction in mouse models and human patients with AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Córtex Entorrinal/patologia , Hipocampo/patologia , Rede Nervosa/patologia , Neurônios/efeitos dos fármacos , Sinapses/patologia , Doença de Alzheimer/psicologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Transtornos Cognitivos/psicologia , Progressão da Doença , Eletroencefalografia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Placa Amiloide/patologia
5.
J Neurophysiol ; 104(4): 2214-23, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20631216

RESUMO

The density of somatostatin (SOM)-containing GABAergic interneurons in the hilus of the dentate gyrus is significantly decreased in both human and experimental temporal lobe epilepsy. We used the pilocarpine model of status epilepticus and temporal lobe epilepsy in mice to study anatomical and electrophysiological properties of surviving somatostatin interneurons and determine whether compensatory functional changes occur that might offset loss of other inhibitory neurons. Using standard patch-clamp techniques and pipettes containing biocytin, whole cell recordings were obtained in hippocampal slices maintained in vitro. Hilar SOM cells containing enhanced green fluorescent protein (EGFP) were identified with fluorescent and infrared differential interference contrast video microscopy in epileptic and control GIN (EGFP-expressing Inhibitory Neurons) mice. Results showed that SOM cells from epileptic mice had 1) significant increases in somatic area and dendritic length; 2) changes in membrane properties, including a small but significant decrease in resting membrane potential, and increases in time constant and whole cell capacitance; 3) increased frequency of slowly rising spontaneous excitatory postsynaptic currents (sEPSCs) due primarily to increased mEPSC frequency, without changes in the probability of release; 4) increased evoked EPSC amplitude; and 5) increased spontaneous action potential generation in cell-attached recordings. Results suggest an increase in excitatory innervation, perhaps on distal dendrites, considering the slower rising EPSCs and increased output of hilar SOM cells in this model of epilepsy. In sum, these changes would be expected to increase the inhibitory output of surviving SOM interneurons and in part compensate for interneuronal loss in the epileptogenic hippocampus.


Assuntos
Giro Denteado/metabolismo , Modelos Animais de Doenças , Epilepsia/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Interneurônios/metabolismo , Somatostatina/biossíntese , Animais , Doença Crônica , Giro Denteado/química , Giro Denteado/patologia , Epilepsia/patologia , Feminino , Interneurônios/química , Interneurônios/patologia , Masculino , Camundongos , Técnicas de Cultura de Órgãos
6.
J Neurosci ; 30(1): 372-81, 2010 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20053918

RESUMO

Previous studies suggested that cleavage of the amyloid precursor protein (APP) at aspartate residue 664 by caspases may play a key role in the pathogenesis of Alzheimer's disease. Mutation of this site (D664A) prevents caspase cleavage and the generation of the C-terminal APP fragments C31 and Jcasp, which have been proposed to mediate amyloid-beta (Abeta) neurotoxicity. Here we compared human APP transgenic mice with (B254) and without (J20) the D664A mutation in a battery of tests. Before Abeta deposition, hAPP-B254 and hAPP-J20 mice had comparable hippocampal levels of Abeta(1-42). At 2-3 or 5-7 months of age, hAPP-B254 and hAPP-J20 mice had similar abnormalities relative to nontransgenic mice in spatial and nonspatial learning and memory, elevated plus maze performance, electrophysiological measures of synaptic transmission and plasticity, and levels of synaptic activity-related proteins. Thus, caspase cleavage of APP at position D664 and generation of C31 do not play a critical role in the development of these abnormalities.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Caspases/metabolismo , Modelos Animais de Doenças , Neurônios/metabolismo , Desempenho Psicomotor/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Humanos , Hidrólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neurônios/patologia
7.
Cell Stem Cell ; 5(6): 624-33, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19951690

RESUMO

Adult neurogenesis regulates plasticity and function in the hippocampus, which is critical for memory and vulnerable to Alzheimer's disease (AD). Promoting neurogenesis may improve hippocampal function in AD brains. However, how amyloid beta (Abeta), the key AD pathogen, affects the development and function of adult-born neurons remains unknown. Adult-born granule cells (GCs) in human amyloid precursor protein (hAPP) transgenic mice, an AD model, showed greater dendritic length, spine density, and functional responses than did controls early in development, but were impaired morphologically and functionally during later maturation. Early inhibition of GABA(A) receptors to suppress GABAergic signaling or late inhibition of calcineurin to enhance glutamatergic signaling normalized the development of adult-born GCs in hAPP mice with high Abeta levels. Abeta-induced increases in GABAergic neurotransmission or an imbalance between GABAergic and glutamatergic neurotransmission may contribute to impaired neurogenesis in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Inibidores de Calcineurina , Diferenciação Celular/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Antagonistas de Receptores de GABA-A , Hipocampo/efeitos dos fármacos , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Humanos , Camundongos , Camundongos Transgênicos , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neurogênese/efeitos dos fármacos , Toxina Pertussis/administração & dosagem , Transmissão Sináptica/efeitos dos fármacos , Tacrolimo/administração & dosagem
8.
Cell Stem Cell ; 5(6): 634-45, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19951691

RESUMO

Apolipoprotein (apo) E, a polymorphic protein with three isoforms (apoE2, apoE3, and apoE4), is essential for lipid homeostasis. Carriers of apoE4 are at higher risk for developing Alzheimer's disease. We have investigated adult neurogenesis in mice with knockout (KO) for apoE or with knockin (KI) alleles for human apoE3 or apoE4, and we report that neurogenesis is reduced in both apoE-KO and apoE4-KI mice. In apoE-KO mice, increased BMP signaling promoted glial differentiation at the expense of neurogenesis. In contrast, in apoE4-KI mice, presynaptic GABAergic input-mediated maturation of newborn neurons was diminished. Tau phosphorylation, an Alzheimer's disease characteristic, and levels of neurotoxic apoE fragments were both elevated in apoE4-KI hippocampal neurons concomitant with decreased GABAergic interneuron survival. Potentiating GABAergic signaling restored neuronal maturation and neurogenesis in apoE4-KI mice to normal levels. These findings suggest that GABAergic signaling can be targeted to mitigate the deleterious effects of apoE4 on neurogenesis.


Assuntos
Células-Tronco Adultas/metabolismo , Doença de Alzheimer/metabolismo , Apolipoproteínas/metabolismo , Neuroglia/metabolismo , Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Animais Recém-Nascidos , Apolipoproteínas/genética , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Agonistas GABAérgicos/administração & dosagem , Técnicas de Introdução de Genes , Hipocampo/patologia , Humanos , Interneurônios/metabolismo , Interneurônios/patologia , Camundongos , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas tau/metabolismo
9.
Nat Neurosci ; 11(11): 1311-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18931664

RESUMO

Neuronal expression of familial Alzheimer's disease-mutant human amyloid precursor protein (hAPP) and hAPP-derived amyloid-beta (Abeta) peptides causes synaptic dysfunction, inflammation and abnormal cerebrovascular tone in transgenic mice. Fatty acids may be involved in these processes, but their contribution to Alzheimer's disease pathogenesis is uncertain. We used a lipidomics approach to generate a broad profile of fatty acids in brain tissues of hAPP-expressing mice and found an increase in arachidonic acid and its metabolites, suggesting increased activity of the group IV isoform of phospholipase A(2) (GIVA-PLA(2)). The levels of activated GIVA-PLA(2) in the hippocampus were increased in individuals with Alzheimer's disease and in hAPP mice. Abeta caused a dose-dependent increase in GIVA-PLA(2) phosphorylation in neuronal cultures. Inhibition of GIVA-PLA(2) diminished Abeta-induced neurotoxicity. Genetic ablation or reduction of GIVA-PLA(2) protected hAPP mice against Abeta-dependent deficits in learning and memory, behavioral alterations and premature mortality. Inhibition of GIVA-PLA(2) may be beneficial in the treatment and prevention of Alzheimer's disease.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos/enzimologia , Transtornos Cognitivos/etiologia , Fosfolipases A2 do Grupo IV/deficiência , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Ácidos Araquidônicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Encéfalo/citologia , Encéfalo/patologia , Estudos de Casos e Controles , Morte Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Ácidos Graxos/metabolismo , Feminino , Fosfolipases A2 do Grupo IV/metabolismo , Humanos , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Ratos , Receptores de AMPA/metabolismo
10.
Neuron ; 60(2): 247-57, 2008 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-18957217

RESUMO

Impaired degradation of amyloid beta (Abeta) peptides could lead to Abeta accumulation, an early trigger of Alzheimer's disease (AD). How Abeta-degrading enzymes are regulated remains largely unknown. Cystatin C (CysC, CST3) is an endogenous inhibitor of cysteine proteases, including cathepsin B (CatB), a recently discovered Abeta-degrading enzyme. A CST3 polymorphism is associated with an increased risk of late-onset sporadic AD. Here, we identified CysC as the key inhibitor of CatB-induced Abeta degradation in vivo. Genetic ablation of CST3 in hAPP-J20 mice significantly lowered soluble Abeta levels, the relative abundance of Abeta1-42, and plaque load. CysC removal also attenuated Abeta-associated cognitive deficits and behavioral abnormalities and restored synaptic plasticity in the hippocampus. Importantly, the beneficial effects of CysC reduction were abolished on a CatB null background, providing direct evidence that CysC regulates soluble Abeta and Abeta-associated neuronal deficits through inhibiting CatB-induced Abeta degradation.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Catepsina B/metabolismo , Cistatina C/genética , Cistatina C/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiopatologia , Predisposição Genética para Doença/genética , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Polimorfismo Genético/genética
11.
J Neurophysiol ; 96(2): 834-45, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16707715

RESUMO

Classification of inhibitory interneurons is critical in determining their role in normal information processing and pathophysiological conditions such as epilepsy. Classification schemes have relied on morphological, physiological, biochemical, and molecular criteria; and clear correlations have been demonstrated between firing patterns and cellular markers such as neuropeptides and calcium-binding proteins. This molecular diversity has allowed generation of transgenic mouse strains in which GFP expression is linked to the expression of one of these markers and presumably a single subtype of neuron. In the GIN mouse (EGFP-expressing Inhibitory Neurons), a subpopulation of somatostatin-containing interneurons in the hippocampus and neocortex is labeled with enhanced green fluorescent protein (EGFP). To optimize the use of the GIN mouse, it is critical to know whether the population of somatostatin-EGFP-expressing interneurons is homogeneous. We performed unsupervised cluster analysis on 46 EGFP-expressing interneurons, based on data obtained from whole cell patch-clamp recordings. Cells were classified according to a number of electrophysiological variables related to spontaneous excitatory postsynaptic currents (sEPSCs), firing behavior, and intrinsic membrane properties. EGFP-expressing interneurons were heterogeneous and at least four subgroups could be distinguished. In addition, multiple discriminant analysis was applied to data collected during whole cell recordings to develop an algorithm for predicting the group membership of newly encountered EGFP-expressing interneurons. Our data are consistent with a heterogeneous population of neurons based on electrophysiological properties and indicate that EGFP expression in the GIN mouse is not restricted to a single class of somatostatin-positive interneuron.


Assuntos
Interneurônios/classificação , Interneurônios/fisiologia , Córtex Motor/fisiologia , Córtex Somatossensorial/fisiologia , Somatostatina/fisiologia , Algoritmos , Animais , Contagem de Células , Análise por Conglomerados , Estimulação Elétrica , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Modelos Neurológicos , Córtex Motor/citologia , Neocórtex/fisiologia , Técnicas de Patch-Clamp , Córtex Somatossensorial/citologia
12.
J Neurophysiol ; 90(2): 644-54, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12711716

RESUMO

Recurrent and lateral inhibition play a prominent role in patterning the odor-evoked discharges in mitral cells, the output neurons of the olfactory bulb. Inhibitory responses in this brain region are mediated through reciprocal synaptic connections made between the dendrites of mitral cells and GABAergic interneurons. Previous studies have demonstrated that N-methyl-D-aspartate (NMDA) receptors on interneurons play a critical role in eliciting GABA release at reciprocal dendrodendritic synapses. In acute olfactory bulb slices, these receptors are tonically blocked by extracellular Mg2+, and recurrent inhibition is disabled. In the present study, we examined the mechanisms by which this tonic blockade could be reversed. We demonstrate that near-coincident activation of an excitatory pathway to the proximal dendrites of GABAergic interneurons relieves the Mg2+ blockade of NMDA receptors at reciprocal dendrodendritic synapses and greatly facilitates recurrent inhibition onto mitral cells. Gating of recurrent and lateral inhibition in the presence of extracellular Mg2+ requires gamma-frequency stimulation of glutamatergic axons in the granule cell layer. Long-range excitatory axon connections from mitral cells innervated by different subpopulations of olfactory receptor neurons may provide a gating input to granule cells, thereby facilitating the mitral cell lateral inhibition that contributes to odorant encoding.


Assuntos
Dendritos/fisiologia , Potenciais Pós-Sinápticos Excitadores , Interneurônios/fisiologia , Inibição Neural , Bulbo Olfatório/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Estimulação Elétrica , Eletrofisiologia , Magnésio/metabolismo , Bulbo Olfatório/efeitos dos fármacos , Periodicidade , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/agonistas , Receptores de AMPA/fisiologia , Receptores de GABA-A/fisiologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Ácido gama-Aminobutírico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...