Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 598(7879): 144-150, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33184512

RESUMO

Cortical neurons exhibit extreme diversity in gene expression as well as in morphological and electrophysiological properties1,2. Most existing neural taxonomies are based on either transcriptomic3,4 or morpho-electric5,6 criteria, as it has been technically challenging to study both aspects of neuronal diversity in the same set of cells7. Here we used Patch-seq8 to combine patch-clamp recording, biocytin staining, and single-cell RNA sequencing of more than 1,300 neurons in adult mouse primary motor cortex, providing a morpho-electric annotation of almost all transcriptomically defined neural cell types. We found that, although broad families of transcriptomic types (those expressing Vip, Pvalb, Sst and so on) had distinct and essentially non-overlapping morpho-electric phenotypes, individual transcriptomic types within the same family were not well separated in the morpho-electric space. Instead, there was a continuum of variability in morphology and electrophysiology, with neighbouring transcriptomic cell types showing similar morpho-electric features, often without clear boundaries between them. Our results suggest that neuronal types in the neocortex do not always form discrete entities. Instead, neurons form a hierarchy that consists of distinct non-overlapping branches at the level of families, but can form continuous and correlated transcriptomic and morpho-electrical landscapes within families.


Assuntos
Perfilação da Expressão Gênica , Córtex Motor/citologia , Neurônios/classificação , Neurônios/metabolismo , Transcriptoma , Animais , Atlas como Assunto , Feminino , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Glutamatos/metabolismo , Lisina/análogos & derivados , Lisina/análise , Masculino , Camundongos , Córtex Motor/anatomia & histologia , Neurônios/citologia , Especificidade de Órgãos , Técnicas de Patch-Clamp , Fenótipo , Análise de Sequência de RNA , Análise de Célula Única , Coloração e Rotulagem
2.
Proc Natl Acad Sci U S A ; 117(14): 8143-8153, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32209671

RESUMO

Although major depressive disorder (MDD) is highly prevalent, its pathophysiology is poorly understood. Recent evidence suggests that glycogen-synthase kinase 3ß (GSK3ß) plays a key role in memory formation, yet its role in mood regulation remains controversial. Here, we investigated whether GSK3ß activity in the nucleus accumbens (NAc) is associated with depression-like behaviors and synaptic plasticity. We performed whole-cell patch-clamp recordings of medium spiny neurons (MSNs) in the NAc and determined the role of GSK3ß in spike timing-dependent long-term potentiation (tLTP) in the chronic unpredictable mild stress (CUMS) mouse model of depression. To assess the specific role of GSK3ß in tLTP, we used in vivo genetic silencing by an adeno-associated viral vector (AAV2) short hairpin RNA against GSK3ß. In addition, we examined the role of the voltage-gated potassium Kv4.2 subunit, a molecular determinant of A-type K+ currents, as a potential downstream target of GSK3ß. We found increased levels of active GSK3ß and augmented tLTP in CUMS mice, a phenotype that was prevented by selective GSK3ß knockdown. Furthermore, knockdown of GSK3ß in the NAc ameliorated depressive-like behavior in CUMS mice. Electrophysiological, immunohistochemical, biochemical, and pharmacological experiments revealed that inhibition of the Kv4.2 channel through direct phosphorylation at Ser-616 mediated the GSK3ß-dependent tLTP changes in CUMS mice. Our results identify GSK3ß regulation of Kv4.2 channels as a molecular mechanism of MSN maladaptive plasticity underlying depression-like behaviors and suggest that the GSK3ß-Kv4.2 axis may be an attractive therapeutic target for MDD.


Assuntos
Transtorno Depressivo Maior/patologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Plasticidade Neuronal , Núcleo Accumbens/patologia , Canais de Potássio Shal/metabolismo , Potenciais de Ação , Animais , Comportamento Animal , Transtorno Depressivo Maior/etiologia , Transtorno Depressivo Maior/psicologia , Modelos Animais de Doenças , Masculino , Camundongos , Neurônios/patologia , Núcleo Accumbens/citologia , Técnicas de Patch-Clamp , Estresse Psicológico/complicações , Estresse Psicológico/psicologia , Fatores de Tempo
3.
J Neurosci ; 41(6): 1191-1206, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33328293

RESUMO

The dentate gyrus (DG) controls information flow into the hippocampus and is critical for learning, memory, pattern separation, and spatial coding, while DG dysfunction is associated with neuropsychiatric disorders. Despite its importance, the molecular mechanisms regulating DG neural circuit assembly and function remain unclear. Here, we identify the Rac-GEF Tiam1 as an important regulator of DG development and associated memory processes. In the hippocampus, Tiam1 is predominantly expressed in the DG throughout life. Global deletion of Tiam1 in male mice results in DG granule cells with simplified dendritic arbors, reduced dendritic spine density, and diminished excitatory synaptic transmission. Notably, DG granule cell dendrites and synapses develop normally in Tiam1 KO mice, resembling WT mice at postnatal day 21 (P21), but fail to stabilize, leading to dendrite and synapse loss by P42. These results indicate that Tiam1 promotes DG granule cell dendrite and synapse stabilization late in development. Tiam1 loss also increases the survival, but not the production, of adult-born DG granule cells, possibly because of greater circuit integration as a result of decreased competition with mature granule cells for synaptic inputs. Strikingly, both male and female mice lacking Tiam1 exhibit enhanced contextual fear memory and context discrimination. Together, these results suggest that Tiam1 is a key regulator of DG granule cell stabilization and function within hippocampal circuits. Moreover, based on the enhanced memory phenotype of Tiam1 KO mice, Tiam1 may be a potential target for the treatment of disorders involving memory impairments.SIGNIFICANCE STATEMENT The dentate gyrus (DG) is important for learning, memory, pattern separation, and spatial navigation, and its dysfunction is associated with neuropsychiatric disorders. However, the molecular mechanisms controlling DG formation and function remain elusive. By characterizing mice lacking the Rac-GEF Tiam1, we demonstrate that Tiam1 promotes the stabilization of DG granule cell dendritic arbors, spines, and synapses, whereas it restricts the survival of adult-born DG granule cells, which compete with mature granule cells for synaptic integration. Notably, mice lacking Tiam1 also exhibit enhanced contextual fear memory and context discrimination. These findings establish Tiam1 as an essential regulator of DG granule cell development, and identify it as a possible therapeutic target for memory enhancement.


Assuntos
Dendritos/metabolismo , Giro Denteado/metabolismo , Memória/fisiologia , Neurogênese/fisiologia , Sinapses/metabolismo , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T/deficiência , Animais , Dendritos/genética , Giro Denteado/citologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Sinapses/genética , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T/genética
4.
Cereb Cortex ; 29(5): 1851-1865, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29790931

RESUMO

Spike timing-dependent plasticity (STDP) is a form of activity-dependent remodeling of synaptic strength that underlies memory formation. Despite its key role in dictating learning rules in the brain circuits, the molecular mechanisms mediating STDP are still poorly understood. Here, we show that spike timing-dependent long-term depression (tLTD) and A-type K+ currents are modulated by pharmacological agents affecting the levels of active glycogen-synthase kinase 3 (GSK3) and by GSK3ß knockdown in layer 2/3 of the mouse somatosensory cortex. Moreover, the blockade of A-type K+ currents mimics the effects of GSK3 up-regulation on tLTD and occludes further changes in synaptic strength. Pharmacological, immunohistochemical and biochemical experiments revealed that GSK3ß influence over tLTD induction is mediated by direct phosphorylation at Ser-616 of the Kv4.2 subunit, a molecular determinant of A-type K+ currents. Collectively, these results identify the functional interaction between GSK3ß and Kv4.2 channel as a novel mechanism for tLTD modulation providing exciting insight into the understanding of GSK3ß role in synaptic plasticity.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Neurônios/fisiologia , Canais de Potássio Shal/metabolismo , Córtex Somatossensorial/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fosforilação , Córtex Somatossensorial/metabolismo
5.
FASEB J ; 30(6): 2171-86, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26917740

RESUMO

Recent data shows that fibroblast growth factor 14 (FGF14) binds to and controls the function of the voltage-gated sodium (Nav) channel with phenotypic outcomes on neuronal excitability. Mutations in the FGF14 gene in humans have been associated with brain disorders that are partially recapitulated in Fgf14(-/-) mice. Thus, signaling pathways that modulate the FGF14:Nav channel interaction may be important therapeutic targets. Bioluminescence-based screening of small molecule modulators of the FGF14:Nav1.6 complex identified 4,5,6,7 -: tetrabromobenzotriazole (TBB), a potent casein kinase 2 (CK2) inhibitor, as a strong suppressor of FGF14:Nav1.6 interaction. Inhibition of CK2 through TBB reduces the interaction of FGF14 with Nav1.6 and Nav1.2 channels. Mass spectrometry confirmed direct phosphorylation of FGF14 by CK2 at S228 and S230, and mutation to alanine at these sites modified FGF14 modulation of Nav1.6-mediated currents. In 1 d in vitro hippocampal neurons, TBB induced a reduction in FGF14 expression, a decrease in transient Na(+) current amplitude, and a hyperpolarizing shift in the voltage dependence of Nav channel steady-state inactivation. In mature neurons, TBB reduces the axodendritic polarity of FGF14. In cornu ammonis area 1 hippocampal slices from wild-type mice, TBB impairs neuronal excitability by increasing action potential threshold and lowering firing frequency. Importantly, these changes in excitability are recapitulated in Fgf14(-/-) mice, and deletion of Fgf14 occludes TBB-dependent phenotypes observed in wild-type mice. These results suggest that a CK2-FGF14 axis may regulate Nav channels and neuronal excitability.-Hsu, W.-C. J., Scala, F., Nenov, M. N., Wildburger, N. C., Elferink, H., Singh, A. K., Chesson, C. B., Buzhdygan, T., Sohail, M., Shavkunov, A. S., Panova, N. I., Nilsson, C. L., Rudra, J. S., Lichti, C. F., Laezza, F. CK2 activity is required for the interaction of FGF14 with voltage-gated sodium channels and neuronal excitability.


Assuntos
Caseína Quinase II/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Neurônios/fisiologia , Canais de Sódio Disparados por Voltagem/fisiologia , Animais , Caseína Quinase II/genética , Feminino , Fatores de Crescimento de Fibroblastos/genética , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Hipocampo/citologia , Hipocampo/fisiologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp
6.
J Neurosci ; 34(38): 12893-903, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25232124

RESUMO

Intracellular accumulation of amyloid-ß (Aß) protein has been proposed as an early event in AD pathogenesis. In patients with mild cognitive impairment, intraneuronal Aß immunoreactivity was found especially in brain regions critically involved in the cognitive deficits of AD. Although a large body of evidence demonstrates that Aß42 accumulates intraneuronally ((in)Aß), the action and the role of Aß42 buildup on synaptic function have been poorly investigated. Here, we demonstrate that basal synaptic transmission and LTP were markedly depressed following Aß42 injection into the neuron through the patch pipette. Control experiments performed with the reverse peptide (Aß42-1) allowed us to exclude that the effects of (in)Aß depended on changes in oncotic pressure. To further investigate (in)Aß synaptotoxicity we used an Aß variant harboring oxidized methionine in position 35 that does not cross the neuronal plasma membrane and is not uploaded from the extracellular space. This Aß42 variant had no effects on synaptic transmission and plasticity when applied extracellularly, but induced synaptic depression and LTP inhibition after patch-pipette dialysis. Finally, the injection of an antibody raised against human Aß42 (6E10) in CA1 pyramidal neurons of mouse hippocampal brain slices and autaptic microcultures did not, per se, significantly affect LTP and basal synaptic transmission, but it protected against the toxic effects of extracellular Aß42. Collectively, these findings suggest that Aß42-induced impairment of glutamatergic synaptic function depends on its internalization and intracellular accumulation thus paving the way to a systemic proteomic analysis of intracellular targets/partners of Aß42.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Ácido Glutâmico/fisiologia , Hipocampo/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Peptídeos beta-Amiloides/administração & dosagem , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/fisiologia , Espaço Intracelular/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Microinjeções , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/administração & dosagem , Cultura Primária de Células , Transmissão Sináptica/fisiologia
8.
Nat Commun ; 13(1): 6389, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36302912

RESUMO

Neocortical feedback is critical for attention, prediction, and learning. To mechanically understand its function requires deciphering its cell-type wiring. Recent studies revealed that feedback between primary motor to primary somatosensory areas in mice is disinhibitory, targeting vasoactive intestinal peptide-expressing interneurons, in addition to pyramidal cells. It is unknown whether this circuit motif represents a general cortico-cortical feedback organizing principle. Here we show that in contrast to this wiring rule, feedback between higher-order lateromedial visual area to primary visual cortex preferentially activates somatostatin-expressing interneurons. Functionally, both feedback circuits temporally sharpen feed-forward excitation eliciting a transient increase-followed by a prolonged decrease-in pyramidal cell activity under sustained feed-forward input. However, under feed-forward transient input, the primary motor to primary somatosensory cortex feedback facilitates bursting while lateromedial area to primary visual cortex feedback increases time precision. Our findings argue for multiple cortico-cortical feedback motifs implementing different dynamic non-linear operations.


Assuntos
Interneurônios , Células Piramidais , Camundongos , Animais , Retroalimentação , Interneurônios/fisiologia , Peptídeo Intestinal Vasoativo
9.
Front Mol Neurosci ; 14: 643860, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276302

RESUMO

The axon initial segment (AIS) is a highly regulated subcellular domain required for neuronal firing. Changes in the AIS protein composition and distribution are a form of structural plasticity, which powerfully regulates neuronal activity and may underlie several neuropsychiatric and neurodegenerative disorders. Despite its physiological and pathophysiological relevance, the signaling pathways mediating AIS protein distribution are still poorly studied. Here, we used confocal imaging and whole-cell patch clamp electrophysiology in primary hippocampal neurons to study how AIS protein composition and neuronal firing varied in response to selected kinase inhibitors targeting the AKT/GSK3 pathway, which has previously been shown to phosphorylate AIS proteins. Image-based features representing the cellular pattern distribution of the voltage-gated Na+ (Nav) channel, ankyrin G, ßIV spectrin, and the cell-adhesion molecule neurofascin were analyzed, revealing ßIV spectrin as the most sensitive AIS protein to AKT/GSK3 pathway inhibition. Within this pathway, inhibition of AKT by triciribine has the greatest effect on ßIV spectrin localization to the AIS and its subcellular distribution within neurons, a phenotype that Support Vector Machine classification was able to accurately distinguish from control. Treatment with triciribine also resulted in increased excitability in primary hippocampal neurons. Thus, perturbations to signaling mechanisms within the AKT pathway contribute to changes in ßIV spectrin distribution and neuronal firing that may be associated with neuropsychiatric and neurodegenerative disorders.

10.
Elife ; 92020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32134385

RESUMO

Clones of excitatory neurons derived from a common progenitor have been proposed to serve as elementary information processing modules in the neocortex. To characterize the cell types and circuit diagram of clonally related excitatory neurons, we performed multi-cell patch clamp recordings and Patch-seq on neurons derived from Nestin-positive progenitors labeled by tamoxifen induction at embryonic day 10.5. The resulting clones are derived from two radial glia on average, span cortical layers 2-6, and are composed of a random sampling of transcriptomic cell types. We find an interaction between shared lineage and connection type: related neurons are more likely to be connected vertically across cortical layers, but not laterally within the same layer. These findings challenge the view that related neurons show uniformly increased connectivity and suggest that integration of vertical intra-clonal input with lateral inter-clonal input may represent a developmentally programmed connectivity motif supporting the emergence of functional circuits.


Assuntos
Neocórtex/citologia , Neurônios/classificação , Neurônios/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Camundongos
11.
Nat Commun ; 10(1): 4174, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31519874

RESUMO

Layer 4 (L4) of mammalian neocortex plays a crucial role in cortical information processing, yet a complete census of its cell types and connectivity remains elusive. Using whole-cell recordings with morphological recovery, we identified one major excitatory and seven inhibitory types of neurons in L4 of adult mouse visual cortex (V1). Nearly all excitatory neurons were pyramidal and all somatostatin-positive (SOM+) non-fast-spiking interneurons were Martinotti cells. In contrast, in somatosensory cortex (S1), excitatory neurons were mostly stellate and SOM+ interneurons were non-Martinotti. These morphologically distinct SOM+ interneurons corresponded to different transcriptomic cell types and were differentially integrated into the local circuit with only S1 neurons receiving local excitatory input. We propose that cell type specific circuit motifs, such as the Martinotti/pyramidal and non-Martinotti/stellate pairs, are used across the cortex as building blocks to assemble cortical circuits.


Assuntos
Neocórtex/citologia , Animais , Eletrofisiologia , Feminino , Interneurônios/citologia , Interneurônios/metabolismo , Masculino , Camundongos , Neocórtex/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/metabolismo , Somatostatina/metabolismo
13.
ACS Chem Neurosci ; 9(5): 976-987, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29359916

RESUMO

Protein-protein interactions (PPI) offer unexploited opportunities for CNS drug discovery and neurochemical probe development. Here, we present ZL181, a novel peptidomimetic targeting the PPI interface of the voltage-gated Na+ channel Nav1.6 and its regulatory protein fibroblast growth factor 14 (FGF14). ZL181 binds to FGF14 and inhibits its interaction with the Nav1.6 channel C-tail. In HEK-Nav1.6 expressing cells, ZL181 acts synergistically with FGF14 to suppress Nav1.6 current density and to slow kinetics of fast inactivation, but antagonizes FGF14 modulation of steady-state inactivation that is regulated by the N-terminal tail of the protein. In medium spiny neurons in the nucleus accumbens, ZL181 suppresses excitability by a mechanism that is dependent upon expression of FGF14 and is consistent with a state-dependent inhibition of FGF14. Overall, ZL181 and derivatives could lay the ground for developing allosteric modulators of Nav channels that are of interest for a broad range of CNS disorders.


Assuntos
Fatores de Crescimento de Fibroblastos/farmacologia , Hipocampo/efeitos dos fármacos , Sódio/metabolismo , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Animais , Fatores de Crescimento de Fibroblastos/genética , Células HEK293 , Humanos , Camundongos Knockout , Peptidomiméticos/farmacologia
14.
Cell Rep ; 23(2): 555-567, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642012

RESUMO

Resilience and vulnerability to neuropsychiatric disorders are linked to molecular changes underlying excitability that are still poorly understood. Here, we identify glycogen-synthase kinase 3ß (GSK3ß) and voltage-gated Na+ channel Nav1.6 as regulators of neuroplasticity induced by environmentally enriched (EC) or isolated (IC) conditions-models for resilience and vulnerability. Transcriptomic studies in the nucleus accumbens from EC and IC rats predicted low levels of GSK3ß and SCN8A mRNA as a protective phenotype associated with reduced excitability in medium spiny neurons (MSNs). In vivo genetic manipulations demonstrate that GSK3ß and Nav1.6 are molecular determinants of MSN excitability and that silencing of GSK3ß prevents maladaptive plasticity of IC MSNs. In vitro studies reveal direct interaction of GSK3ß with Nav1.6 and phosphorylation at Nav1.6T1936 by GSK3ß. A GSK3ß-Nav1.6T1936 competing peptide reduces MSNs excitability in IC, but not EC rats. These results identify GSK3ß regulation of Nav1.6 as a biosignature of MSNs maladaptive plasticity.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Condicionamento Físico Animal , Isolamento Social , Animais , Potenciais Evocados , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/genética , Células HEK293 , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.6/química , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Técnicas de Patch-Clamp , Fosfopeptídeos/análise , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transcriptoma
15.
Nat Protoc ; 12(12): 2531-2553, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29189773

RESUMO

Neurons exhibit a rich diversity of morphological phenotypes, electrophysiological properties, and gene-expression patterns. Understanding how these different characteristics are interrelated at the single-cell level has been difficult because of the lack of techniques for multimodal profiling of individual cells. We recently developed Patch-seq, a technique that combines whole-cell patch-clamp recording, immunohistochemistry, and single-cell RNA-sequencing (scRNA-seq) to comprehensively profile single neurons from mouse brain slices. Here, we present a detailed step-by-step protocol, including modifications to the patching mechanics and recording procedure, reagents and recipes, procedures for immunohistochemistry, and other tips to assist researchers in obtaining high-quality morphological, electrophysiological, and transcriptomic data from single neurons. Successful implementation of Patch-seq allows researchers to explore the multidimensional phenotypic variability among neurons and to correlate gene expression with phenotype at the level of single cells. The entire procedure can be completed in ∼2 weeks through the combined efforts of a skilled electrophysiologist, molecular biologist, and biostatistician.


Assuntos
Perfilação da Expressão Gênica/métodos , Imuno-Histoquímica/métodos , Neurônios/citologia , Técnicas de Patch-Clamp/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Animais , Células Cultivadas , Fenômenos Eletrofisiológicos , Camundongos , Neurônios/metabolismo , Transcriptoma
16.
Front Cell Neurosci ; 11: 225, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28804449

RESUMO

Hippocampal plasticity is triggered by a variety of stimuli including sensory inputs, neurotrophins and inflammation. Leptin, whose primary function is to regulate food intake and energy expenditure, has been recently shown to affect hippocampal neurogenesis and plasticity. Interestingly, mice fed a high-fat diet (HFD) exhibit impaired hippocampal function, but the underlying mechanisms are poorly understood. To address this issue, we compared leptin responsiveness of hippocampal neurons in control and HFD-fed mice by combining single-cell electrophysiology and biochemical assays. We found that leptin modulated spontaneous and evoked synaptic transmission in control, but not HFD, mice. This functional impairment was paralleled by blunted activation of STAT-3, one of the key signal transduction pathways controlled by the fully functional isoform of the leptin receptor, ObRb. In addition, SOCS-3 expression was non-responsive to leptin, indicating that modulation of negative feedback impinging on ObRb was also altered. Our results advance the understanding of leptin action on hippocampal plasticity and, more importantly, suggest that leptin resistance is a key determinant of hippocampal dysfunction associated with hypercaloric diet.

17.
Neuropharmacology ; 117: 49-60, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28126496

RESUMO

Psychiatric disorders such as anxiety, depression and addiction are often comorbid brain pathologies thought to share common mechanistic biology. As part of the cortico-limbic circuit, the nucleus accumbens shell (NAcSh) plays a fundamental role in integrating information in the circuit, such that modulation of NAcSh circuitry alters anxiety, depression, and addiction-related behaviors. Intracellular kinase cascades in the NAcSh have proven important mediators of behavior. To investigate glycogen-synthase kinase 3 (GSK3) beta signaling in the NAcSh in vivo we knocked down GSK3beta expression with a novel adeno-associated viral vector (AAV2) and assessed changes in anxiety- and depression-like behavior and cocaine self-administration in GSK3beta knockdown rats. GSK3beta knockdown reduced anxiety-like behavior while increasing depression-like behavior and cocaine self-administration. Correlative electrophysiological recordings in acute brain slices were used to assess the effect of AAV-shGSK3beta on spontaneous firing and intrinsic excitability of tonically active interneurons (TANs), cells required for input and output signal integration in the NAcSh and for processing reward-related behaviors. Loose-patch recordings showed that TANs transduced by AAV-shGSK3beta exhibited reduction in tonic firing and increased spike half width. When assessed by whole-cell patch clamp recordings these changes were mirrored by reduction in action potential firing and accompanied by decreased hyperpolarization-induced depolarizing sag potentials, increased action potential current threshold, and decreased maximum rise time. These results suggest that silencing of GSK3beta in the NAcSh increases depression- and addiction-related behavior, possibly by decreasing intrinsic excitability of TANs. However, this study does not rule out contributions from other neuronal sub-types.


Assuntos
Ansiedade/genética , Comportamento Aditivo/genética , Comportamento Animal/fisiologia , Depressão/genética , Glicogênio Sintase Quinase 3 beta/fisiologia , Interneurônios/fisiologia , Núcleo Accumbens/fisiologia , Potenciais de Ação/fisiologia , Animais , Cocaína/farmacologia , Técnicas de Silenciamento de Genes , Glicogênio Sintase Quinase 3 beta/genética , Masculino , Ratos , Autoadministração
18.
Nat Commun ; 8(1): 2009, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29222408

RESUMO

High-fat diet (HFD) and metabolic diseases cause detrimental effects on hippocampal synaptic plasticity, learning, and memory through molecular mechanisms still poorly understood. Here, we demonstrate that HFD increases palmitic acid deposition in the hippocampus and induces hippocampal insulin resistance leading to FoxO3a-mediated overexpression of the palmitoyltransferase zDHHC3. The excess of palmitic acid along with higher zDHHC3 levels causes hyper-palmitoylation of AMPA glutamate receptor subunit GluA1, hindering its activity-dependent trafficking to the plasma membrane. Accordingly, AMPAR current amplitudes and, more importantly, their potentiation underlying synaptic plasticity were inhibited, as well as hippocampal-dependent memory. Hippocampus-specific silencing of Zdhhc3 and, interestingly enough, intranasal injection of the palmitoyltransferase inhibitor, 2-bromopalmitate, counteract GluA1 hyper-palmitoylation and restore synaptic plasticity and memory in HFD mice. Our data reveal a key role of FoxO3a/Zdhhc3/GluA1 axis in the HFD-dependent impairment of cognitive function and identify a novel mechanism underlying the cross talk between metabolic and cognitive disorders.


Assuntos
Encéfalo/fisiologia , Proteína Forkhead Box O3/metabolismo , Hipocampo/fisiologia , Resistência à Insulina , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de AMPA/metabolismo , Animais , Células Cultivadas , Cognição/fisiologia , Dieta Hiperlipídica , Lipoilação , Potenciação de Longa Duração/fisiologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Interferência de RNA
19.
Mol Neurobiol ; 53(10): 7254-7270, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26687232

RESUMO

Adult neurogenesis, the production of mature neurons from progenitor cells in the adult mammalian brain, is linked to the etiology of neurodegenerative and psychiatric disorders. However, a thorough understanding of the molecular elements at the base of adult neurogenesis remains elusive. Here, we provide evidence for a previously undescribed function of fibroblast growth factor 14 (FGF14), a brain disease-associated factor that controls neuronal excitability and synaptic plasticity, in regulating adult neurogenesis in the dentate gyrus (DG). We found that FGF14 is dynamically expressed in restricted subtypes of sex determining region Y-box 2 (Sox2)-positive and doublecortin (DCX)-positive neural progenitors in the DG. Bromodeoxyuridine (BrdU) incorporation studies and confocal imaging revealed that genetic deletion of Fgf14 in Fgf14 -/- mice leads to a significant change in the proportion of proliferating and immature and mature newly born adult granule cells. This results in an increase in the late immature and early mature population of DCX and calretinin (CR)-positive neurons. Electrophysiological extracellular field recordings showed reduced minimal threshold response and impaired paired-pulse facilitation at the perforant path to DG inputs in Fgf14 -/- compared to Fgf14 +/+ mice, supporting disrupted synaptic connectivity as a correlative read-out to impaired neurogenesis. These new insights into the biology of FGF14 in neurogenesis shed light into the signaling pathways associated with disrupted functions in complex brain diseases.


Assuntos
Envelhecimento/metabolismo , Grânulos Citoplasmáticos/metabolismo , Giro Denteado/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Neurogênese , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Contagem de Células , Diferenciação Celular , Sobrevivência Celular , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Fatores de Crescimento de Fibroblastos/genética , Deleção de Genes , Perfilação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/citologia , Neuropeptídeos/metabolismo , Sinapses/metabolismo
20.
Neurobiol Aging ; 36(2): 886-900, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25541422

RESUMO

Amyloid ß-protein (Aß) pathologies have been linked to dysfunction of excitability in neurons of the hippocampal circuit, but the molecular mechanisms underlying this process are still poorly understood. Here, we applied whole-cell patch-clamp electrophysiology to primary hippocampal neurons and show that intracellular Aß42 delivery leads to increased spike discharge and action potential broadening through downregulation of A-type K(+) currents. Pharmacologic studies showed that caspases and glycogen synthase kinase 3 (GSK-3) activation are required for these Aß42-induced effects. Extracellular perfusion and subsequent internalization of Aß42 increase spike discharge and promote GSK-3-dependent phosphorylation of the Kv4.2 α-subunit, a molecular determinant of A-type K(+) currents, at Ser-616. In acute hippocampal slices derived from an adult triple-transgenic Alzheimer's mouse model, characterized by endogenous intracellular accumulation of Aß42, CA1 pyramidal neurons exhibit hyperexcitability accompanied by increased phosphorylation of Kv4.2 at Ser-616. Collectively, these data suggest that intraneuronal Aß42 accumulation leads to an intracellular cascade culminating into caspases activation and GSK-3-dependent phosphorylation of Kv4.2 channels. These findings provide new insights into the toxic mechanisms triggered by intracellular Aß42 and offer potentially new therapeutic targets for Alzheimer's disease treatment.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/toxicidade , Caspases/fisiologia , Fenômenos Eletrofisiológicos/genética , Quinase 3 da Glicogênio Sintase/fisiologia , Hipocampo/fisiopatologia , Neurônios/fisiologia , Fragmentos de Peptídeos/toxicidade , Canais de Potássio Shal/metabolismo , Potenciais de Ação/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Hipocampo/citologia , Hipocampo/metabolismo , Técnicas In Vitro , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Terapia de Alvo Molecular , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosforilação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA