Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 132024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38748470

RESUMO

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Assuntos
Neurônios Colinérgicos , Dopamina , Interneurônios , Optogenética , Dopamina/metabolismo , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/fisiologia , Ratos , Optogenética/métodos , Motivação , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Acetilcolina/metabolismo
2.
Cell Rep ; 43(3): 113834, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38431842

RESUMO

Striatal dopamine axons co-release dopamine and gamma-aminobutyric acid (GABA), using GABA provided by uptake via GABA transporter-1 (GAT1). Functions of GABA co-release are poorly understood. We asked whether co-released GABA autoinhibits dopamine release via axonal GABA type A receptors (GABAARs), complementing established inhibition by dopamine acting at axonal D2 autoreceptors. We show that dopamine axons express α3-GABAAR subunits in mouse striatum. Enhanced dopamine release evoked by single-pulse optical stimulation in striatal slices with GABAAR antagonism confirms that an endogenous GABA tone limits dopamine release. Strikingly, an additional inhibitory component is seen when multiple pulses are used to mimic phasic axonal activity, revealing the role of GABAAR-mediated autoinhibition of dopamine release. This autoregulation is lost in conditional GAT1-knockout mice lacking GABA co-release. Given the faster kinetics of ionotropic GABAARs than G-protein-coupled D2 autoreceptors, our data reveal a mechanism whereby co-released GABA acts as a first responder to dampen phasic-to-tonic dopamine signaling.


Assuntos
Autorreceptores , Dopamina , Camundongos , Animais , Ácido gama-Aminobutírico/farmacologia , Axônios/metabolismo , Corpo Estriado/metabolismo , Receptores de GABA-A/metabolismo , Camundongos Knockout , Homeostase
3.
bioRxiv ; 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38260459

RESUMO

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. A recent paper in eLife (Mohebi et al., 2023) reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1. Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.

4.
Mol Neurobiol ; 60(7): 4105-4119, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37022587

RESUMO

The ability to store, retrieve, and extinguish memories of adverse experiences is an essential skill for animals' survival. The cellular and molecular factors that underlie such processes are only partially known. Using chondroitinase ABC treatment targeting chondroitin sulfate proteoglycans (CSPGs), previous studies showed that the maturation of the extracellular matrix makes fear memory resistant to deletion. Mice lacking the cartilage link protein Crtl1 (Crtl1-KO mice) display normal CSPG levels but impaired CSPG condensation in perineuronal nets (PNNs). Thus, we asked whether the presence of PNNs in the adult brain is responsible for the appearance of persistent fear memories by investigating fear extinction in Crtl1-KO mice. We found that mutant mice displayed fear memory erasure after an extinction protocol as revealed by analysis of freezing and pupil dynamics. Fear memory erasure did not depend on passive loss of retention; moreover, we demonstrated that, after extinction training, conditioned Crtl1-KO mice display no neural activation in the amygdala (Zif268 staining) in comparison to control animals. Taken together, our findings suggest that the aggregation of CSPGs into PNNs regulates the boundaries of the critical period for fear extinction.


Assuntos
Extinção Psicológica , Proteínas da Matriz Extracelular , Medo , Animais , Camundongos , Encéfalo/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo
5.
Cell Rep ; 39(3): 110716, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35443174

RESUMO

Dopamine (DA)-releasing neurons in the substantia nigra pars compacta (SNcDA) inhibit target cells in the striatum through postsynaptic activation of γ-aminobutyric acid (GABA) receptors. However, the molecular mechanisms responsible for GABAergic signaling remain unclear, as SNcDA neurons lack enzymes typically required to produce GABA or package it into synaptic vesicles. Here, we show that aldehyde dehydrogenase 1a1 (Aldh1a1), an enzyme proposed to function as a GABA synthetic enzyme in SNcDA neurons, does not produce GABA for synaptic transmission. Instead, we demonstrate that SNcDA axons obtain GABA exclusively through presynaptic uptake using the membrane GABA transporter Gat1 (encoded by Slc6a1). GABA is then packaged for vesicular release using the vesicular monoamine transporter Vmat2. Our data therefore show that presynaptic transmitter recycling can substitute for de novo GABA synthesis and that Vmat2 contributes to vesicular GABA transport, expanding the range of molecular mechanisms available to neurons to support inhibitory synaptic communication.


Assuntos
Dopamina , Neurônios Dopaminérgicos , Corpo Estriado , Mesencéfalo , Substância Negra/fisiologia , Ácido gama-Aminobutírico
6.
Nat Metab ; 3(3): 293-294, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33758418
7.
Biol Psychiatry ; 86(9): 682-692, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31235076

RESUMO

BACKGROUND: Rodents and humans show an attenuation of fear extinction during adolescence, which coincides with the onset of several psychiatric disorders. Although the ethological relevance and the underlying mechanism are largely unknown, the suppression of fear extinction during adolescence is associated with a diminished plasticity in the glutamatergic neurons of the infralimbic medial prefrontal cortex, a brain region critical for fear extinction. Given the putative effect of synaptic inhibition on glutamatergic neuron activity, we studied whether gamma-aminobutyric acidergic neurons in the infralimbic medial prefrontal cortex are involved in the suppression of fear extinction during adolescence. METHODS: We assessed membrane and synaptic properties in parvalbumin-positive interneurons (PVINs) and somatostatin-positive interneurons (SSTINs) in male preadolescent, adolescent, and adult mice. The effect of fear conditioning and extinction on PVIN-pyramidal neuron and SSTIN-pyramidal neuron synapses in male preadolescent, adolescent, and adult mice was evaluated using an optogenetic approach. RESULTS: The development of the membrane excitability of PVINs is delayed and reaches maturity only by adulthood, while the SSTIN membrane properties are developed early and remain stable during development from preadolescence to adulthood. Although the synaptic inhibition mediated by PVINs undergoes a protracted development, it does not exhibit a fear behavior-specific plasticity. However, the synaptic inhibition mediated by SSTINs undergoes an adolescence-specific enhancement, and this increased inhibition is suppressed by fear learning but is not restored by extinction training. This altered plasticity during adolescence overlapped with a reduction in calcium-permeable glutamate receptors in SSTINs. CONCLUSIONS: The adolescence-specific plasticity in the SSTINs might play a role in fear extinction suppression during adolescence in mice.


Assuntos
Extinção Psicológica , Interneurônios/fisiologia , Sistema Límbico/crescimento & desenvolvimento , Plasticidade Neuronal , Córtex Pré-Frontal/crescimento & desenvolvimento , Animais , Medo , Inibição Psicológica , Sistema Límbico/citologia , Sistema Límbico/fisiologia , Masculino , Camundongos , Optogenética , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Sinapses/fisiologia
8.
Neuropsychopharmacology ; 44(10): 1828-1836, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31005058

RESUMO

The medial habenula-interpeduncular nucleus (MHb-IPN) pathway has recently been implicated in the suppression of fear memory. A notable feature of this pathway is the corelease of neurotransmitters and neuropeptides from MHb neurons. Our studies in mice reveal that an activation of substance P-positive dorsomedial habenula (dMHb) neurons results in simultaneous release of glutamate and glycine in the lateral interpeduncular nucleus (LIPN). This glycine receptor activity inhibits an activity-dependent long-lasting potentiation of glutamatergic synapses in LIPN neurons, while substance P enhances this plasticity. An endocannabinoid CB1 receptor-mediated suppression of GABAB receptor activity allows substance P to induce a long-lasting increase in glutamate release in LIPN neurons. Consistent with the substance P-dependent synaptic potentiation in the LIPN, the NK1R in the IPN is involved in fear extinction but not fear conditioning. Thus, our study describes a novel plasticity mechanism in the LIPN and a region-specific role of substance P in fear extinction.


Assuntos
Glicina/metabolismo , Habenula/metabolismo , Núcleo Interpeduncular/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Substância P/metabolismo , Animais , Fenômenos Eletrofisiológicos , Ácido Glutâmico/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Potenciação de Longa Duração/fisiologia , Camundongos , Receptor CB1 de Canabinoide/metabolismo , Receptores de GABA-B/metabolismo , Receptores da Neurocinina-1/metabolismo , Transmissão Sináptica
9.
eNeuro ; 5(5)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30255129

RESUMO

Rett syndrome (RTT) is caused in most cases by loss-of-function mutations in the X-linked gene encoding methyl CpG-binding protein 2 (MECP2). Understanding the pathological processes impacting sensory-motor control represents a major challenge for clinical management of individuals affected by RTT, but the underlying molecular and neuronal modifications remain unclear. We find that symptomatic male Mecp2 knockout (KO) mice show atypically elevated parvalbumin (PV) expression in both somatosensory (S1) and motor (M1) cortices together with excessive excitatory inputs converging onto PV-expressing interneurons (INs). In accordance, high-speed voltage-sensitive dye imaging shows reduced amplitude and spatial spread of synaptically induced neuronal depolarizations in S1 of Mecp2 KO mice. Moreover, motor learning-dependent changes of PV expression and structural synaptic plasticity typically occurring on PV+ INs in M1 are impaired in symptomatic Mecp2 KO mice. Finally, we find similar abnormalities of PV networks plasticity in symptomatic female Mecp2 heterozygous mice. These results indicate that in Mecp2 mutant mice the configuration of PV+ INs network is shifted toward an atypical plasticity state in relevant cortical areas compatible with the sensory-motor dysfunctions characteristics of RTT.


Assuntos
Interneurônios/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Plasticidade Neuronal/fisiologia , Parvalbuminas/metabolismo , Síndrome de Rett/metabolismo , Animais , Modelos Animais de Doenças , Camundongos Knockout , Neurônios/metabolismo , Sinapses/metabolismo
10.
Exp Cell Res ; 368(2): 225-235, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29730163

RESUMO

Mutations in MECP2 gene have been identified in more than 95% of patients with classic Rett syndrome, one of the most common neurodevelopmental disorders in females. Taking advantage of the breakthrough technology of genetic reprogramming, we investigated transcriptome changes in neurons differentiated from induced Pluripotent Stem Cells (iPSCs) derived from patients with different mutations. Profiling by RNA-seq in terminally differentiated neurons revealed a prominent GABAergic circuit disruption along with a perturbation of cytoskeleton dynamics. In particular, in mutated neurons we identified a significant decrease of acetylated α-tubulin which can be reverted by treatment with selective inhibitors of HDAC6, the main α-tubulin deacetylase. These findings contribute to shed light on Rett pathogenic mechanisms and provide hints for the treatment of Rett-associated epileptic behavior as well as for the definition of new therapeutic strategies for Rett syndrome.


Assuntos
Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Desacetilase 6 de Histona/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatologia , Tubulina (Proteína)/metabolismo , Acetilação , Diferenciação Celular/fisiologia , Feminino , Humanos , Masculino
11.
Mol Neurobiol ; 55(12): 9267-9279, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29663284

RESUMO

Hypothalamic proopiomelanocortin (POMC) neurons are important players in the regulation of energy homeostasis; we previously demonstrated that environmental stimulation excites arcuate nucleus circuits to undergo plastic remodeling, leading to altered ratio between excitatory and inhibitory synaptic contacts on these neurons. The widely used selective serotonin reuptake inhibitor fluoxetine (FLX) is known to affect body weight. On the other hand, FLX administration mimics the effects of environmental stimulation on synaptic plasticity in the hippocampus and cortex. The mammalian target of rapamycin (mTOR) pathway is instrumental in these phenomena. Thus, we aimed at investigating whether and how FLX affects POMC neurons activity and hypothalamic mTOR function. Adult mice expressing green fluorescent protein (GFP) under the POMC promoter were treated with FLX for 3 weeks resulting in diminished body weight. Patch clamp recordings performed on POMC neurons indicate that FLX increases their firing rate and the excitatory AMPA-mediated transmission, and reduces the inhibitory GABAergic currents at presynaptic level. Immunofluorescence studies indicate that FLX increases the ratio between excitatory and inhibitory synaptic contacts on POMC neurons. These changes are associated with an increased activity of the hypothalamic mTOR pathway. Use of the mTOR inhibitor rapamycin blunts the effects of FLX on body weight and on functional and structural plasticity of POMC neurons. Our findings indicate that FLX is able to remodel POMC neurons, and that this may be partly mediated by the mTOR signaling pathway.


Assuntos
Fluoxetina/farmacologia , Hipotálamo/citologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais , Potenciais de Ação/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Leptina/farmacologia , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Sirolimo/farmacologia
12.
Cell Rep ; 20(5): 1111-1122, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768196

RESUMO

The medial habenula-interpeduncular nucleus (MHb-IPN) pathway, which connects the limbic forebrain to the midbrain, has recently been implicated in aversive behaviors. The MHb-IPN circuit is characterized by a unique topographical organization, an excitatory role of GABA, and a prominent co-release of neurotransmitters and neuropeptides. However, little is known about synaptic plasticity in this pathway. An application of a high-frequency stimulation resulted in a long-lasting potentiation of glutamate release in IPN neurons. Our experiments reveal that a Ca2+-permeable AMPA receptor (CPAR)-dependent release of GABA from IPN neurons and a retrograde activation of GABAB receptors on MHb terminals result in a long-lasting enhancement of glutamate release. Strikingly, adolescent IPN neurons lacked CPARs and exhibited an inability to undergo plasticity. In addition, fear conditioning suppressed an activity-dependent potentiation of MHb-IPN synapses, whereas fear extinction reversed this plasticity deficit, suggesting a role of the MHb-IPN synaptic plasticity in the regulation of aversive behaviors.


Assuntos
Núcleo Interpeduncular/metabolismo , Plasticidade Neuronal/fisiologia , Receptores de AMPA/metabolismo , Receptores de GABA-B/metabolismo , Sinapses/metabolismo , Animais , Núcleo Interpeduncular/citologia , Camundongos , Camundongos Transgênicos , Receptores de AMPA/genética , Receptores de GABA-B/genética , Sinapses/genética
13.
Neuroscience ; 352: 296-305, 2017 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-28412502

RESUMO

Since Ebbinghaus' classical work on oblivion and saving effects, we know that declarative memories may become at first spontaneously irretrievable and only subsequently completely extinguished. Recently, this time-dependent path toward memory-trace loss has been shown to correlate with different patterns of brain activation. Environmental enrichment (EE) enhances learning and memory and affects system memory consolidation. However, there is no evidence on whether and how EE could affect the time-dependent path toward oblivion. We used Object Recognition Test (ORT) to assess in adult mice put in EE for 40days (EE mice) or left in standard condition (SC mice) memory retrieval of the familiar objects 9 and 21days after learning with or without a brief retraining performed the day before. We found that SC mice show preferential exploration of new object at day 9 only with retraining, while EE mice do it even without. At day 21 SC mice do not show preferential exploration of novel object, irrespective of the retraining, while EE mice are still capable to benefit from retraining, even if they were not able to spontaneously recover the trace. Analysis of c-fos expression 20days after learning shows a different pattern of active brain areas in response to the retraining session in EE and SC mice, with SC mice recruiting the same brain network as naïve SC or EE mice following de novo learning. This suggests that EE promotes formation of longer lasting object recognition memory, allowing a longer time window during which saving is present.


Assuntos
Meio Ambiente , Reconhecimento Psicológico/fisiologia , Análise de Variância , Animais , Encéfalo/metabolismo , Comportamento Exploratório/fisiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Masculino , Rememoração Mental/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/metabolismo , Aprendizagem Espacial/fisiologia , Fatores de Tempo
14.
Neuropharmacology ; 113(Pt A): 167-177, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-26924708

RESUMO

Environmental enrichment (EE) has a remarkable impact on brain development. Continuous exposure to EE from birth determines a significant acceleration of visual system maturation both at retinal and cortical levels. A pre-weaning enriched experience is sufficient to trigger the accelerated maturation of the visual system, suggesting that factors affected by EE during the first days of life might prime visual circuits towards a faster development. The search for such factors is crucial not only to gain a better understanding of the molecular hierarchy of brain development but also to identify molecular pathways amenable to be targeted to correct atypical brain developmental trajectories. Here, we showed that IGF-1 levels are increased in the visual cortex of EE rats as early as P6 and this is a crucial event for setting in motion the developmental program induced by EE. Early intracerebroventricular (i.c.v.) infusion of IGF-1 in standard rats was sufficient to mimic the action of EE on visual acuity development, whereas blocking IGF-1 signaling by i.c.v. injections of the IGF-1 receptor antagonist JB1 prevented the deployment of EE effects. Early IGF-1 decreased the ratio between the expression of NKCC1 and KCC2 cation/chloride transporters, and the reversal potential for GABAAR-driven Cl- currents (ECl) was shifted toward more negative potentials, indicating that IGF-1 is a crucial factor in accelerating the maturation of GABAergic neurotransmission and promoting the developmental switch of GABA polarity from excitation to inhibition. In addition, early IGF-1 promoted a later occurring increase in its own expression, suggesting a priming effect of early IGF-1 in driving post-weaning cortical maturation.


Assuntos
Fator de Crescimento Insulin-Like I/fisiologia , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Simportadores/metabolismo , Córtex Visual/crescimento & desenvolvimento , Córtex Visual/fisiologia , Animais , Meio Ambiente , Neurônios GABAérgicos/fisiologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/metabolismo , Parvalbuminas/metabolismo , Ratos , Ratos Long-Evans , Receptores de GABA/fisiologia , Acuidade Visual/fisiologia , Córtex Visual/metabolismo , Cotransportadores de K e Cl-
15.
Neuron ; 92(4): 873-887, 2016 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-27746130

RESUMO

Glial cells respond to neuronal activation and release neuroactive molecules (termed "gliotransmitters") that can affect synaptic activity and modulate plasticity. In this study, we used molecular genetic tools, ultra-structural microscopy, and electrophysiology to assess the role of brain-derived neurotrophic factor (BDNF) on cortical gliotransmission in vivo. We find that glial cells recycle BDNF that was previously secreted by neurons as pro-neurotrophin following long-term potentiation (LTP)-inducing electrical stimulation. Upon BDNF glial recycling, we observed tight, temporal, highly localized TrkB phosphorylation on adjacent neurons, a process required to sustain LTP. Engagement of BDNF recycling by astrocytes represents a novel mechanism by which cortical synapses can expand BDNF action and provide synaptic changes that are relevant for the acquisition of new memories. Accordingly, mice deficient in BDNF glial recycling fail to recognize familiar from novel objects, indicating a physiological requirement for this process in memory consolidation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Potenciação de Longa Duração , Memória , Neuroglia/metabolismo , Receptor trkB/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Sinapses/metabolismo , Animais , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Camundongos , Fosforilação , Receptores de Fator de Crescimento Neural/genética
16.
Biol Psychiatry ; 80(4): 302-311, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26452614

RESUMO

BACKGROUND: CDKL5 (cyclin-dependent kinase-like 5) is mutated in many severe neurodevelopmental disorders, including atypical Rett syndrome. CDKL5 was shown to interact with synaptic proteins, but an in vivo analysis of the role of CDKL5 in dendritic spine dynamics and synaptic molecular organization is still lacking. METHODS: In vivo two-photon microscopy of the somatosensory cortex of Cdkl5(-/y) mice was applied to monitor structural dynamics of dendritic spines. Synaptic function and plasticity were measured using electrophysiological recordings of excitatory postsynaptic currents and long-term potentiation in brain slices and assessing the expression of synaptic postsynaptic density protein 95 (PSD-95). Finally, we studied the impact of insulin-like growth factor 1 (IGF-1) treatment on CDKL5 null mice to restore the synaptic deficits. RESULTS: Adult mutant mice showed a significant reduction in spine density and PSD-95-positive synaptic puncta, a reduction of persistent spines, and impaired long-term potentiation. In juvenile mutants, short-term spine elimination, but not formation, was dramatically increased. Exogenous administration of IGF-1 rescued defective rpS6 phosphorylation, spine density, and PSD-95 expression. Endogenous cortical IGF-1 levels were unaffected by CDKL5 deletion. CONCLUSIONS: These data demonstrate that dendritic spine stabilization is strongly regulated by CDKL5. Moreover, our data suggest that IGF-1 treatment could be a promising candidate for clinical trials in CDKL5 patients.


Assuntos
Espinhas Dendríticas/patologia , Fator de Crescimento Insulin-Like I/uso terapêutico , Síndrome de Rett , Espasmos Infantis , Fatores Etários , Animais , Animais Recém-Nascidos , Espinhas Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Síndromes Epilépticas , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Guanilato Quinases/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Células Piramidais/patologia , Células Piramidais/ultraestrutura , Síndrome de Rett/tratamento farmacológico , Síndrome de Rett/genética , Síndrome de Rett/patologia , Córtex Somatossensorial/patologia , Espasmos Infantis/tratamento farmacológico , Espasmos Infantis/genética , Espasmos Infantis/patologia , Sinapses/efeitos dos fármacos , Sinapses/patologia
17.
Exp Neurol ; 265: 48-58, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25483398

RESUMO

Alzheimer's disease is the most prevalent tauopathy and cause of dementia. We investigate the hypothesis that reactivation of plasticity can restore function in the presence of neuronal damage resulting from tauopathy. We investigated two models with tau hyperphosphorylation, aggregation and neurodegeneration: a transgenic mouse model in which the mutant P301S tau is expressed in neurons (Tg P301S), and a model in which an adeno-associated virus expressing P301S tau (AAV-P301S) was injected in the perirhinal cortex, a region critical for object recognition (OR) memory. Both models show profound loss of OR memory despite only 15% neuronal loss in the Tg P301S and 26% in AAV-P301S-injected mice. Recordings from perirhinal cortex slices of 3month-old P301S transgenic mice showed a diminution in synaptic transmission following temporal stimulation. Chondroitinase ABC (ChABC) can reactivate plasticity and affect memory through actions on perineuronal nets. ChABC was injected into the perirhinal cortex and animals were tested for OR memory 1week later, demonstrating restoration of OR memory to normal levels. Synaptic transmission indicated by fEPSP amplitude was restored to control levels following ChABC treatment. ChABC did not affect the progression of neurodegenerative tauopathy. These findings suggest that increasing plasticity by manipulation of perineuronal nets offers a novel therapeutic approach to the treatment of memory loss in neurodegenerative disorders.


Assuntos
Córtex Cerebral/enzimologia , Condroitina ABC Liase/administração & dosagem , Memória/fisiologia , Rede Nervosa/enzimologia , Plasticidade Neuronal/fisiologia , Tauopatias/enzimologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Humanos , Injeções Intraventriculares , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/patologia , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Tauopatias/tratamento farmacológico , Tauopatias/patologia
18.
J Neurosci ; 34(4): 1542-53, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24453341

RESUMO

A major challenge in the neuroscience field is the identification of molecules and pathways that control synaptic plasticity and memory. Dendritic spines play a pivotal role in these processes, as the major sites of excitatory synapses in neuronal communication. Previous studies have shown that the scaffold protein p140Cap localizes into dendritic spines and that its knockdown negatively modulates spine shape in culture. However, so far, there is no information on its in vivo relevance. By using a knock-out mouse model, we here demonstrate that p140Cap is a key element for both learning and synaptic plasticity. Indeed, p140Cap(-/-) mice are impaired in object recognition test, as well as in LTP and in LTD measurements. The in vivo effects of p140Cap loss are presumably attenuated by noncell-autonomous events, since primary neurons obtained from p140Cap(-/-) mice show a strong reduction in number of mushroom spines and abnormal organization of synapse-associated F-actin. These phenotypes are most likely caused by a local reduction of the inhibitory control of RhoA and of cortactin toward the actin-depolymerizing factor cofilin. These events can be controlled by p140Cap through its capability to directly inhibit the activation of Src kinase and by its binding to the scaffold protein Citron-N. Altogether, our results provide new insight into how protein associated with dynamic microtubules may regulate spine actin organization through interaction with postsynaptic density components.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinases da Família src/metabolismo , Actinas/metabolismo , Animais , Western Blotting , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Imunofluorescência , Hipocampo/metabolismo , Aprendizagem/fisiologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Ratos , Transdução de Sinais/fisiologia , Transmissão Sináptica/fisiologia
19.
J Neurosci ; 33(16): 7057-65, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595763

RESUMO

Perineuronal nets (PNNs) are extracellular matrix structures surrounding cortical neuronal cell bodies and proximal dendrites and are involved in the control of brain plasticity and the closure of critical periods. Expression of the link protein Crtl1/Hapln1 in neurons has recently been identified as the key event triggering the formation of PNNs. Here we show that the genetic attenuation of PNNs in adult brain Crtl1 knock-out mice enhances long-term object recognition memory and facilitates long-term depression in the perirhinal cortex, a neural correlate of object recognition memory. Identical prolongation of memory follows localized digestion of PNNs with chondroitinase ABC, an enzyme that degrades the chondroitin sulfate proteoglycan components of PNNs. The memory-enhancing effect of chondroitinase ABC treatment attenuated over time, suggesting that the regeneration of PNNs gradually restored control plasticity levels. Our findings indicate that PNNs regulate both memory and experience-driven synaptic plasticity in adulthood.


Assuntos
Córtex Cerebral/fisiologia , Matriz Extracelular/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Neurônios/fisiologia , Reconhecimento Psicológico/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Condroitina ABC Liase/farmacologia , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas da Matriz Extracelular/deficiência , Genótipo , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Penicilinase/farmacologia , Proteoglicanas/deficiência , Reconhecimento Psicológico/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...