Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell ; 149(3): 708-21, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22541439

RESUMO

Alzheimer's disease (AD) results in cognitive decline and altered network activity, but the mechanisms are unknown. We studied human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Electroencephalographic recordings in hAPP mice revealed spontaneous epileptiform discharges, indicating network hypersynchrony, primarily during reduced gamma oscillatory activity. Because this oscillatory rhythm is generated by inhibitory parvalbumin (PV) cells, network dysfunction in hAPP mice might arise from impaired PV cells. Supporting this hypothesis, hAPP mice and AD patients had decreased levels of the interneuron-specific and PV cell-predominant voltage-gated sodium channel subunit Nav1.1. Restoring Nav1.1 levels in hAPP mice by Nav1.1-BAC expression increased inhibitory synaptic activity and gamma oscillations and reduced hypersynchrony, memory deficits, and premature mortality. We conclude that reduced Nav1.1 levels and PV cell dysfunction critically contribute to abnormalities in oscillatory rhythms, network synchrony, and memory in hAPP mice and possibly in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Interneurônios/metabolismo , Aprendizagem , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1 , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Canais de Sódio/metabolismo , Sinapses
2.
Eur J Neurosci ; 59(7): 1558-1566, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38308520

RESUMO

The excitation-inhibition imbalance manifesting as epileptic activities in Alzheimer's disease is gaining more and more attention, and several potentially involved cellular and molecular pathways are currently under investigation. Based on in vitro studies, dopamine D1-type receptors in the anterior cingulate cortex and the hippocampus have been proposed to participate in this peculiar co-morbidity in mouse models of amyloidosis. Here, we tested the implication of dopaminergic transmission in vivo in the Tg2576 mouse model of Alzheimer's disease by monitoring epileptic activities via intracranial EEG before and after treatment with dopamine antagonists. Our results show that neither the D1-like dopamine receptor antagonist SCH23390 nor the D2-like dopamine receptor antagonist haloperidol reduces the frequency of epileptic activities. While requiring further investigation, our results indicate that on a systemic level, dopamine receptors are not significantly contributing to epilepsy observed in vivo in this mouse model of Alzheimer's disease.


Assuntos
Doença de Alzheimer , Amiloidose , Epilepsia , Camundongos , Animais , Antagonistas de Dopamina/farmacologia , Doença de Alzheimer/tratamento farmacológico , Receptores de Dopamina D2/metabolismo , Benzazepinas/farmacologia , Benzazepinas/uso terapêutico , Receptores de Dopamina D1/metabolismo , Epilepsia/tratamento farmacológico , Modelos Animais de Doenças , Amiloidose/tratamento farmacológico
3.
Proc Natl Acad Sci U S A ; 109(42): E2895-903, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-22869752

RESUMO

In light of the rising prevalence of Alzheimer's disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-ß peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-ß-induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anticonvulsivantes/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Cognição/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Piracetam/análogos & derivados , Sinapses/efeitos dos fármacos , Doença de Alzheimer/complicações , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Anticonvulsivantes/sangue , Anticonvulsivantes/uso terapêutico , Western Blotting , Transtornos Cognitivos/etiologia , Eletroencefalografia , Humanos , Imuno-Histoquímica , Levetiracetam , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Rede Nervosa/fisiopatologia , Piracetam/sangue , Piracetam/farmacologia , Piracetam/uso terapêutico
4.
Nat Commun ; 15(1): 4100, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773091

RESUMO

In most models of neuronal plasticity and memory, dopamine is thought to promote the long-term maintenance of Long-Term Potentiation (LTP) underlying memory processes, but not the initiation of plasticity or new information storage. Here, we used optogenetic manipulation of midbrain dopamine neurons in male DAT::Cre mice, and discovered that stimulating the Schaffer collaterals - the glutamatergic axons connecting CA3 and CA1 regions - of the dorsal hippocampus concomitantly with midbrain dopamine terminals within a 200 millisecond time-window triggers LTP at glutamatergic synapses. Moreover, we showed that the stimulation of this dopaminergic pathway facilitates contextual learning in awake behaving mice, while its inhibition hinders it. Thus, activation of midbrain dopamine can operate as a teaching signal that triggers NeoHebbian LTP and promotes supervised learning.


Assuntos
Dopamina , Neurônios Dopaminérgicos , Hipocampo , Aprendizagem , Potenciação de Longa Duração , Optogenética , Área Tegmentar Ventral , Animais , Potenciação de Longa Duração/fisiologia , Área Tegmentar Ventral/fisiologia , Masculino , Dopamina/metabolismo , Camundongos , Neurônios Dopaminérgicos/fisiologia , Neurônios Dopaminérgicos/metabolismo , Hipocampo/fisiologia , Hipocampo/metabolismo , Aprendizagem/fisiologia , Camundongos Transgênicos , Região CA1 Hipocampal/fisiologia , Região CA1 Hipocampal/citologia , Sinapses/fisiologia , Sinapses/metabolismo , Camundongos Endogâmicos C57BL , Memória/fisiologia
5.
Transl Psychiatry ; 13(1): 227, 2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37365183

RESUMO

Mood disorders are associated with hypothalamic-pituitary-adrenal axis overactivity resulting from a decreased inhibitory feedback exerted by the hippocampus on this brain structure. Growing evidence suggests that antidepressants would regulate hippocampal excitatory/inhibitory balance to restore an effective inhibition on this stress axis. While these pharmacological compounds produce beneficial clinical effects, they also have limitations including their long delay of action. Interestingly, non-pharmacological strategies such as environmental enrichment improve therapeutic outcome in depressed patients as in animal models of depression. However, whether exposure to enriched environment also reduces the delay of action of antidepressants remains unknown. We investigated this issue using the corticosterone-induced mouse model of depression, submitted to antidepressant treatment by venlafaxine, alone or in combination with enriched housing. We found that the anxio-depressive phenotype of male mice was improved after only two weeks of venlafaxine treatment when combined with enriched housing, which is six weeks earlier than mice treated with venlafaxine but housed in standard conditions. Furthermore, venlafaxine combined with exposure to enriched environment is associated with a reduction in the number of parvalbumin-positive neurons surrounded by perineuronal nets (PNN) in the mouse hippocampus. We then showed that the presence of PNN in depressed mice prevented their behavioral recovery, while pharmacological degradation of hippocampal PNN accelerated the antidepressant action of venlafaxine. Altogether, our data support the idea that non-pharmacological strategies can shorten the onset of action of antidepressants and further identifies PV interneurons as relevant actors of this effect.


Assuntos
Parvalbuminas , Inibidores Seletivos de Recaptação de Serotonina , Camundongos , Masculino , Animais , Cloridrato de Venlafaxina/farmacologia , Parvalbuminas/metabolismo , Serotonina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Antidepressivos/metabolismo , Interneurônios/metabolismo
6.
Neurobiol Aging ; 123: 35-48, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36634385

RESUMO

The link between Alzheimer's disease (AD) and network hypersynchrony - manifesting as epileptic activities - received considerable attention in the past decade. However, several questions remain unanswered as to its mechanistic underpinnings. Therefore, our objectives were (1) to better characterise epileptic events in the Tg2576 mouse model throughout the sleep-wake cycle and disease progression via electrophysiological recordings and (2) to explore the involvement of noradrenergic transmission in this pathological hypersynchrony. Over and above confirming the previously described early presence and predominance of epileptic events during rapid-eye-movement (REM) sleep, we also show that these events do not worsen with age and are highly phase-locked to the section of the theta cycle during REM sleep where hippocampal pyramidal cells reach their highest firing probability. Finally, we reveal an antiepileptic mechanism of noradrenergic transmission via α1-adrenoreceptors that could explain the intriguing distribution of epileptic events over the sleep-wake cycle in this model, with potential therapeutic implications in the treatment of the epileptic events occurring in many AD patients.


Assuntos
Doença de Alzheimer , Epilepsia , Camundongos , Animais , Doença de Alzheimer/patologia , Camundongos Transgênicos , Sono/fisiologia , Modelos Animais de Doenças , Sono REM
7.
Neuron ; 110(17): 2854-2866.e4, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35858622

RESUMO

Area CA2 is a critical region for diverse hippocampal functions including social recognition memory. This region has unique properties and connectivity. Notably, intra-hippocampal excitatory inputs to CA2 lack canonical long-term plasticity, but inhibitory transmission expresses a long-term depression mediated by Delta-opioid receptors (DOR-iLTDs). Evidence indicates that DOR-iLTDs are insufficient to underlie social coding. Here, we report a novel inhibitory plasticity mediated by cannabinoid type 1 receptor activation (CB1R-iLTD). Surprisingly, CB1R-iLTD requires previous induction of DOR-iLTDs, indicating a permissive role for DOR plasticity. Blockade of CB1Rs in CA2 completely prevents social memory formation. Furthermore, the sequentiality of DOR- and CB1R-mediated plasticity occurs in vivo during successive social interactions. Finally, CB1R-iLTD is altered in a mouse model of schizophrenia with impaired social cognition but is rescued by a manipulation that also rescues social memory. Altogether, our data reveal a unique interplay between two inhibitory plasticities and a novel mechanism for social memory formation.


Assuntos
Hipocampo , Plasticidade Neuronal , Animais , Camundongos , Plasticidade Neuronal/fisiologia , Receptor CB1 de Canabinoide , Reconhecimento Psicológico
8.
iScience ; 25(3): 103895, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35243253

RESUMO

Parvalbumin (PV)-expressing interneurons which are often associated with the specific extracellular matrix perineuronal net (PNN) play a critical role in the alteration of brain activity and memory performance in Alzheimer's disease (AD). The integrity of these neurons is crucial for normal functioning of the hippocampal subfield CA2, and hence, social memory formation. Here, we find that social memory deficits of mouse models of AD are associated with decreased presence of PNN around PV cells and long-term synaptic plasticity in area CA2. Furthermore, single local injection of the growth factor neuregulin-1 (NRG1) is sufficient to restore both PV/PNN levels and social memory performance of these mice. Thus, the PV/PNN disruption in area CA2 could play a causal role in social memory deficits of AD mice, and activating PV cell pro-maturation pathways may be sufficient to restore social memory.

9.
Cell Rep ; 29(5): 1099-1112.e4, 2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31665627

RESUMO

Adolescence is a vulnerable period characterized by major cognitive changes. The mechanisms underlying the emergence of new cognitive functions are poorly understood. We find that a long-term depression of inhibitory transmission (iLTD) from parvalbumin-expressing (PV+) interneurons in the hippocampal area Cornu Ammonis 2 (CA2) is absent in young mice but emerges at the end of adolescence. We demonstrate that the maturation of both the perineuronal net (PNN) and signaling through ErbB4 is required for this plasticity. Furthermore, we demonstrate that social recognition memory displays the same age dependence as iLTD and is impaired by targeted degradation of the PNN or iLTD blockade in area CA2. Our data reveal an unusual developmental rule for plasticity at the PV+ interneuron transmission in area CA2 and indicate that this plasticity is involved in the emergence of higher cognitive function, such as social memory formation, in late adolescence.


Assuntos
Região CA2 Hipocampal/metabolismo , Interneurônios/metabolismo , Memória , Plasticidade Neuronal , Parvalbuminas/metabolismo , Receptor ErbB-4/metabolismo , Transdução de Sinais , Comportamento Social , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Depressão Sináptica de Longo Prazo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Neuregulina-1/metabolismo , Receptores Opioides delta/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo
10.
J Neurosci ; 27(25): 6771-80, 2007 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-17581964

RESUMO

Alzheimer's disease (AD) is characterized by severe neuronal loss in several brain regions important for learning and memory. Of the structures affected by AD, the hippocampus is unique in continuing to produce new neurons throughout life. Mounting evidence indicates that hippocampal neurogenesis contributes to the processing and storage of new information and that deficits in the production of new neurons may impair learning and memory. Here, we examine whether the overproduction of amyloid-beta (Abeta) peptide in a mouse model for AD might be detrimental to newborn neurons in the hippocampus. We used transgenic mice overexpressing familial AD variants of amyloid precursor protein (APP) and/or presenilin-1 to test how the level (moderate or high) and the aggregation state (soluble or deposited) of Abeta impacts the proliferation and survival of new hippocampal neurons. Although proliferation and short-term survival of neural progenitors in the hippocampus was unaffected by APP/Abeta overproduction, survival of newborn cells 4 weeks later was dramatically diminished in transgenic mice with Alzheimer's-type amyloid pathology. Phenotypic analysis of the surviving population revealed a specific reduction in newborn neurons. Our data indicate that overproduction of Abeta and the consequent appearance of amyloid plaques cause an overall reduction in the number of adult-generated hippocampal neurons. Diminished capacity for hippocampal neuron replacement may contribute to the cognitive decline observed in these mice.


Assuntos
Doença de Alzheimer/patologia , Amiloidose/patologia , Hipocampo/citologia , Hipocampo/patologia , Neurônios/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloidose/genética , Amiloidose/metabolismo , Animais , Sobrevivência Celular/genética , Senescência Celular/genética , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo
11.
Behav Brain Res ; 191(1): 104-10, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18433892

RESUMO

Animal models of genetic diseases obtained by transferring human mutated genes in the mouse are widely used in biomedical based research. They constitute efficient tools to study mechanisms underlying abnormal phenotypes. Unfortunately, the phenotype of the transgene is often obscured by the genetic background of the embryonic stem cells and that of the recipient strain used to create the transgenic line. It is also known, from the literature, that repeatedly backcrossing a transgenic strain to an inbred background may have unfavorable effects that can result in the loss of the transgenic line. In order to analyze the influences of the genetic background on the transgene expression, we studied the effects of the hAPPswe transgene involved in Alzheimer's Amyloid Pathology, in 3 genetic backgrounds differing by their genetic heterogeneity (homozygous vs heterozygous) and the strain of origin (C57BL6, CBA, B6SJL F1) after only one generation backcrossing. Three different behavioral paradigms were used to assess the psychological and cognitive phenotypic differences: elevated plus maze, morris navigation task and contextual fear conditioning. Our data indicate that the best solution to maintain the transgenic line is to backcross repeatedly the transgenic mice into the F1 hybrid cross that was used to create the transgenic strain, whereas phenotyping should be performed comparatively after only one generation backcrossing into various well chosen F1 or inbred backgrounds.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Fenótipo , Análise de Variância , Animais , Comportamento Animal/fisiologia , Condicionamento Psicológico/fisiologia , Cruzamentos Genéticos , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Medo , Heterozigoto , Humanos , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação
12.
Neurobiol Aging ; 72: 147-158, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273829

RESUMO

Recent findings show that parvalbumin (PV) interneuron function is impaired in Alzheimer's disease (AD), and that this impairment in PV function can be linked to network dysfunction and memory deficits. PV cells are often associated with a specific extracellular matrix, the perineuronal net (PNN). PNNs are believed to protect PV cell integrity, and whether the amyloidopathy affects PNNs remains unclear. Here, we evaluated the number of PV cells with and without PNNs in the hippocampus of the Tg2576 mouse model of AD at different stages of the disease. We show a deficit of PV+ and/or PV+/PNN+ cells in the areas CA1, CA2, and CA3 in Tg2576 as young as 3 months of age. Importantly, transient exposure to an enriched environment, which has proven long-lasting beneficial effects on memory in AD subjects, rescues the PV/PNN cell number deficits. We conclude that cognitive improvements induced by enriched environment in AD mouse models could be supported by a remodeling of hippocampal PV cell network and their PNNs.


Assuntos
Envelhecimento , Doença de Alzheimer , Meio Ambiente , Matriz Extracelular , Hipocampo/citologia , Interneurônios/citologia , Parvalbuminas/metabolismo , Fatores Etários , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
eNeuro ; 5(6)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30627643

RESUMO

Alzheimer's disease (AD) begins several decades before the onset of clinical symptoms, at a time when women may still undergo reproductive cycling. Whether ovarian functions alter substrates of AD pathogenesis is unknown. Here we show that ovarian cycle stages significantly modulate AD-related alterations in neural network patterns, cognitive impairments, and pathogenic protein production in the hAPP-J20 mouse model of AD. Female hAPP mice spent more time in estrogen-dominant cycle stages and these ovarian stages worsened AD-related network dysfunction and cognitive impairments. In contrast, progesterone-dominant stages and gonadectomy attenuated these AD-related deficits. Further studies revealed a direct role for estradiol in stimulating neural network excitability and susceptibility to seizures in hAPP mice and increasing amyloid beta levels. Understanding dynamic effects of the ovarian cycle on the female nervous system in disease, including AD, is of critical importance and may differ from effects on a healthy brain. The pattern of ovarian cycle effects on disease-related networks, cognition, and pathogenic protein expression may be relevant to young women at risk for AD.


Assuntos
Doença de Alzheimer/complicações , Ondas Encefálicas/fisiologia , Encéfalo/patologia , Transtornos Cognitivos , Ciclo Menstrual/fisiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Castração , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Convulsivantes/toxicidade , Modelos Animais de Doenças , Estradiol/metabolismo , Comportamento Exploratório/fisiologia , Feminino , Humanos , Ciclo Menstrual/genética , Camundongos , Camundongos Transgênicos , Mutação/genética , Pentilenotetrazol/toxicidade , Progesterona/metabolismo , Convulsões/induzido quimicamente , Convulsões/fisiopatologia
14.
Neuron ; 98(1): 75-89.e5, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29551491

RESUMO

Inhibitory interneurons regulate the oscillatory rhythms and network synchrony that are required for cognitive functions and disrupted in Alzheimer's disease (AD). Network dysrhythmias in AD and multiple neuropsychiatric disorders are associated with hypofunction of Nav1.1, a voltage-gated sodium channel subunit predominantly expressed in interneurons. We show that Nav1.1-overexpressing, but not wild-type, interneuron transplants derived from the embryonic medial ganglionic eminence (MGE) enhance behavior-dependent gamma oscillatory activity, reduce network hypersynchrony, and improve cognitive functions in human amyloid precursor protein (hAPP)-transgenic mice, which simulate key aspects of AD. Increased Nav1.1 levels accelerated action potential kinetics of transplanted fast-spiking and non-fast-spiking interneurons. Nav1.1-deficient interneuron transplants were sufficient to cause behavioral abnormalities in wild-type mice. We conclude that the efficacy of interneuron transplantation and the function of transplanted cells in an AD-relevant context depend on their Nav1.1 levels. Disease-specific molecular optimization of cell transplants may be required to ensure therapeutic benefits in different conditions.


Assuntos
Doença de Alzheimer/metabolismo , Ondas Encefálicas/fisiologia , Encéfalo/metabolismo , Cognição/fisiologia , Interneurônios/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.1/biossíntese , Potenciais de Ação/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Animais , Encéfalo/cirurgia , Modelos Animais de Doenças , Expressão Gênica , Hipocampo/metabolismo , Hipocampo/cirurgia , Humanos , Interneurônios/transplante , Locomoção/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1/genética
15.
Psychoneuroendocrinology ; 32 Suppl 1: S26-30, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17629417

RESUMO

It is now widely accepted that new neurons continue to be added to the brain throughout life including during normal aging. The finding of adult neurogenesis in the hippocampus, a structure involved in the processing of memories, has favored the idea that newborn neurons might subserve cognitive functions. Recent work on human post-mortem tissues and mice models of Alzheimer's disease (AD) has reported persistent hippocampal proliferative capacity during pathological aging. Although it is not yet clear whether neurogenesis leads to the production of fully functional mature neurons in AD brains, these findings open prospects for cell-replacement therapies. Strategies aimed at promoting neurogenesis may also contribute to improve cognitive deficits caused by normal or pathological aging.


Assuntos
Envelhecimento/fisiologia , Diferenciação Celular/fisiologia , Demência/patologia , Hipocampo/citologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Demência/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Células-Tronco/metabolismo
17.
J Comp Neurol ; 495(5): 573-86, 2006 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-16498678

RESUMO

Locus coeruleus (LC) noradrenergic neurons are active during wakefulness, slow their discharge rate during slow wave sleep, and stop firing during paradoxical sleep (PS). A large body of data indicates that their inactivation during PS is due to a tonic GABAergic inhibition. To localize the neurons responsible for such inhibition, we first examined the distribution of retrogradely and Fos double-immunostained neurons following cholera toxin b subunit (CTb) injection in the LC of control rats, rats selectively deprived of PS for 3 days, and rats allowed to recover for 3 hours from such deprivation. We found a significant number of CTb/Fos double-labeled cells only in the recovery group. The largest number of CTb/Fos double-labeled cells was found in the dorsal paragigantocellular reticular nucleus (DPGi). It indeed contained 19% of the CTb/Fos double-labeled neurons, whereas the ventrolateral periaqueductal gray (vlPAG) contained 18.3% of these neurons, the lateral paragigantocellular reticular nucleus (LPGi) 15%, the lateral hypothalamic area 9%, the lateral PAG 6.7%, and the rostral PAG 6%. In addition, CTb/Fos double-labeled cells constituted 43% of all the singly CTb-labeled cells counted in the DPGi compared with 29% for the LPGi, 18% for the rostral PAG, and 10% or less for the other structures. Although all these populations of CTb/Fos double-labeled neurons could be GABAergic and tonically inhibit LC neurons during PS, our results indicate that neurons from the DPGi constitute the best candidate for this role.


Assuntos
Locus Cerúleo/citologia , Vias Neurais/anatomia & histologia , Neurônios/metabolismo , Norepinefrina/metabolismo , Sono REM/fisiologia , Animais , Toxina da Cólera/metabolismo , Locus Cerúleo/metabolismo , Masculino , Vias Neurais/metabolismo , Neurônios/citologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Privação do Sono
18.
J Physiol Paris ; 100(5-6): 271-83, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17689057

RESUMO

In the middle of the last century, Michel Jouvet discovered paradoxical sleep (PS), a sleep phase paradoxically characterized by cortical activation and rapid eye movements and a muscle atonia. Soon after, he showed that it was still present in "pontine cats" in which all structures rostral to the brainstem have been removed. Later on, it was demonstrated that the pontine peri-locus coeruleus alpha (peri-LCalpha in cats, corresponding to the sublaterodorsal nucleus, SLD, in rats) is responsible for PS onset. It was then proposed that the onset and maintenance of PS is due to a reciprocal inhibitory interaction between neurons presumably cholinergic specifically active during PS localized in this region and monoaminergic neurons. In the last decade, we have tested this hypothesis with our model of head-restrained rats and functional neuroanatomical studies. Our results confirmed that the SLD in rats contains the neurons responsible for the onset and maintenance of PS. They further indicate that (1) these neurons are non-cholinergic possibly glutamatergic neurons, (2) they directly project to the glycinergic premotoneurons localized in the medullary ventral gigantocellular reticular nucleus (GiV), (3) the main neurotransmitter responsible for their inhibition during waking (W) and slow wave sleep (SWS) is GABA rather than monoamines, (4) they are constantly and tonically excited by glutamate and (5) the GABAergic neurons responsible for their tonic inhibition during W and SWS are localized in the deep mesencephalic reticular nucleus (DPMe). We also showed that the tonic inhibition of locus coeruleus (LC) noradrenergic and dorsal raphe (DRN) serotonergic neurons during sleep is due to a tonic GABAergic inhibition by neurons localized in the dorsal paragigantocellular reticular nucleus (DPGi) and the ventrolateral periaqueductal gray (vlPAG). We propose that these GABAergic neurons also inhibit the GABAergic neurons of the DPMe at the onset and during PS and are therefore responsible for the onset and maintenance of PS.


Assuntos
Acetilcolina/metabolismo , Aminas/metabolismo , Ácido Glutâmico/metabolismo , Modelos Biológicos , Sono REM/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Humanos , Redes Neurais de Computação , Vias Neurais/fisiologia , Neurônios/fisiologia
19.
Brain Struct Funct ; 221(3): 1591-605, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25596866

RESUMO

The neural cell adhesion molecule NCAM and its association with the polysialic acid (PSA) are believed to contribute to brain structural plasticity that underlies memory formation. Indeed, the attachment of long chains of PSA to the glycoprotein NCAM down-regulates its adhesive properties by altering cell-cell interactions. In the brain, the biosynthesis of PSA is catalyzed by two polysialyltransferases, which are differentially regulated during lifespan. One of them, ST8SiaIV (PST), is predominantly expressed during adulthood whereas the other one, ST8SiaII (STX), dominates during embryonic and post-natal development. To understand the role played by ST8SiaIV during learning and memory and its underlying hippocampal plasticity, we used knockout mice deleted for the enzyme ST8SiaIV (PST-ko mice). At adult age, PST-ko mice show a drastic reduction of PSA-NCAM expression in the hippocampus and intact hippocampal adult neurogenesis. We found that these mice display impaired long-term but not short-term memory in both, spatial and non-spatial behavioral tasks. Remarkably, memory deficits of PST-ko mice were abolished by exposure to environmental enrichment that was also associated with an increased number of PSA-NCAM expressing new neurons in the dentate gyrus of these mice. Whether the presence of a larger pool of immature, likely plastic, new neurons favored the rescue of long-term memory in PST-ko mice remains to be determined. Our findings add new evidence to the role played by PSA in memory consolidation. They also suggest that PSA synthesized by PST critically controls the tempo of new neurons maturation in the adult hippocampus.


Assuntos
Ambiente Controlado , Hipocampo/enzimologia , Memória/fisiologia , Moléculas de Adesão de Célula Nervosa/fisiologia , Sialiltransferases/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurogênese , Plasticidade Neuronal , Sialiltransferases/genética , Memória Espacial/fisiologia
20.
Front Aging Neurosci ; 7: 178, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26441640

RESUMO

The cognitive reserve hypothesis claims that the brain can overcome pathology by reinforcing preexistent processes or by developing alternative cognitive strategies. Epidemiological studies have revealed that this reserve can be built throughout life experiences as education or leisure activities. We previously showed that an early transient environmental enrichment (EE) durably improves memory performances in the Tg2576 mouse model of Alzheimer's disease (AD). Recently, we evidenced a hypersynchronous brain network activity in young adult Tg2576 mice. As aberrant oscillatory activity can contribute to memory deficits, we wondered whether the long-lasting memory improvements observed after EE were associated with a reduction of neuronal network hypersynchrony. Thus, we exposed non-transgenic (NTg) and Tg2576 mice to standard or enriched housing conditions for 10 weeks, starting at 3 months of age. Two weeks after EE period, Tg2576 mice presented similar seizure susceptibility to a GABA receptor antagonist. Immediately after and 2 weeks after this enrichment period, standard and enriched-housed Tg2576 mice did not differ with regards to the frequency of interictal spikes on their electroencephalographic (EEG) recordings. Thus, the long-lasting effect of this EE protocol on memory capacities in Tg2576 mice is not mediated by a reduction of their cerebral aberrant neuronal activity at early ages.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA