Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 477
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36835243

RESUMO

Chronic stress is a core risk factor for developing a myriad of neurological disorders, including major depression. The chronicity of such stress can lead to adaptive responses or, on the contrary, to psychological maladaptation. The hippocampus is one of the most affected brain regions displaying functional changes in chronic stress. Egr1, a transcription factor involved in synaptic plasticity, is a key molecule regulating hippocampal function, but its role in stress-induced sequels has been poorly addressed. Emotional and cognitive symptoms were induced in mice by using the chronic unpredictable mild stress (CUMS) protocol. We used inducible double-mutant Egr1-CreERT2 x R26RCE mice to map the formation of Egr1-dependent activated cells. Results show that short- (2 days) or long-term (28 days) stress protocols in mice induce activation or deactivation, respectively, of hippocampal CA1 neural ensembles in an Egr1-activity-dependent fashion, together with an associated dendritic spine pathology. In-depth characterization of these neural ensembles revealed a deep-to-superficial switch in terms of Egr1-dependent activation of CA1 pyramidal neurons. To specifically manipulate deep and superficial pyramidal neurons of the hippocampus, we then used Chrna7-Cre (to express Cre in deep neurons) and Calb1-Cre mice (to express Cre in superficial neurons). We found that specific manipulation of superficial but not deep pyramidal neurons of the CA1 resulted in the amelioration of depressive-like behaviors and the restoration of cognitive impairments induced by chronic stress. In summary, Egr1 might be a core molecule driving the activation/deactivation of hippocampal neuronal subpopulations underlying stress-induced alterations involving emotional and cognitive sequels.


Assuntos
Região CA1 Hipocampal , Cognição , Proteína 1 de Resposta de Crescimento Precoce , Emoções , Células Piramidais , Estresse Psicológico , Animais , Camundongos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia , Doença Crônica , Região CA1 Hipocampal/fisiopatologia
2.
J Neurophysiol ; 128(6): 1566-1577, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36382903

RESUMO

Burst discharges in the immature brain may contribute to its enhanced seizure susceptibility. The cellular mechanisms underlying burst discharges in the CA1 area of the immature versus adult hippocampus were investigated with simultaneous whole-cell and field-potential recordings. When GABAA receptors were blocked pharmacologically, bursts in CA1 were either graded or all-or-none (or mixed) as a function of electrical stimulation intensity. Most CA1 minislices from immature rats displayed all-or-none or mixed bursts, whereas the slices from adult rats predominantly elicited graded bursts. The frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) were greater in CA1 pyramidal cells from the immature than the adult slices. The developmental differences in CA1 bursting were also detected in slices adjusted for maturational changes in brain volume (i.e., 350 µm thick for immature vs. 450 µm thick for adult rats). Neither N-methyl-d-aspartate (NMDA) nor group I metabotropic glutamate (mGlu1) receptor antagonists blocked the network-driven bursts in immature CA1, but an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker abolished them. Robust excitatory postsynaptic potentials (EPSPs) occurred after bursts in some immature CA1 slices (23%) but never in slices from the adult. The input-output (amount of current injected vs. number of action potentials generated) relationship was markedly greater in CA1 pyramidal cells in the immature compared with the adult hippocampus. These data suggest that the CA1 area of the immature brain is capable of generating network-driven bursts, which declines in adult rats. The increased propensity of burst generation in immature CA1 appears to involve a greater AMPA receptor-mediated synaptic network and an increased intrinsic spike-generating ability.NEW & NOTEWORTHY Burst discharges in the developing brain can provide valuable insights into epileptogenesis. We show that the immature hippocampal CA1 area is capable of generating all-or-none (i.e., network) bursts, which transitions to graded (i.e., nonnetwork) bursts in the mature brain via both synaptic and intrinsic mechanisms. Our results provide new clues to help understand possible mechanisms that may be shared in the immature and epileptic brain and how the normal brain becomes seizure prone (i.e., epileptogenesis).


Assuntos
Região CA1 Hipocampal , Convulsões , Animais , Ratos , Região CA1 Hipocampal/fisiologia , Região CA1 Hipocampal/fisiopatologia , Potenciais Pós-Sinápticos Excitadores , Células Piramidais , Fatores Etários
3.
Neuroscience ; 490: 11-24, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35248584

RESUMO

The impacts of early-life adversity (ELA) on cognitive functions including striatal-dependent habit memory and hippocampal-dependent spatial memory were investigated in male mice. The ELA mouse model was generated via an altered cage environment with limited nesting and bedding materials during postnatal days 2-9 (P2-9). The altered cage environment affected the nesting behaviors of dams, creating a stressful condition for their offspring. The ELA mice had biased decision making and poor spatial memory when they grew into young adults (4-month-old). To explore the underlying synaptic basis of these effects, excitatory synapses represented by postsynaptic density protein-95 (PSD-95) were immunolabelled on a series of brain sections and stereologically quantified in the dorsomedial striatum (DMS) and dorsolateral striatum (DLS), as well as in area CA1 of the dorsal hippocampus. Increased PSD-95-immunoreactive synapses were observed in DLS but not DMS, whereas selective loss of PSD-95 synapses was detected in the stratum radiatum of area CA1. The spine data supported the selective effects of ELA on PSD-95 synapses. Specifically, both thin and mushroom-type spines were increased in DLS, while loss of thin spines was apparent in CA1 radiatum in ELA mice versus controls. The correlation between PSD-95 synapses and memory performances was further analyzed, and the data suggested that increased small (<0.20 µm3) and large (>0.40 µm3) synapses in DLS might drive ELA mice to make decisions largely relying on habit memory, while loss of small synapses in hippocampal CA1 damage the spatial memory of ELA mice.


Assuntos
Região CA1 Hipocampal , Espinhas Dendríticas , Memória , Estresse Psicológico , Envelhecimento/psicologia , Animais , Região CA1 Hipocampal/fisiopatologia , Proteína 4 Homóloga a Disks-Large , Masculino , Camundongos , Sinapses
4.
J Stroke Cerebrovasc Dis ; 31(3): 106241, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34983004

RESUMO

OBJECTIVES: Global cerebral ischemia (CI) causes severe neuronal injury, mainly in the hippocampal CA1 region. This study aimed to investigate an immediate using transcranial direct current stimulation (tDCS) in reducing neuronal injury induced by CI. MATERIALS AND METHODS: The 32 Wistar male rats were randomly divided into four groups (n=8 per group). In the ischemia group (I), CI was induced via the 4-vessel occlusion model. In the sham group (Sh), rats did not receive any intervention. In the ischemia+cathodal group (I+c/tDCS), the cathodal current was applied during CI. In the ischemia+anodal group (I+a/tDCS), the anodal current was applied. The current intensity of 400 µA was applied for 15-min during the ischemia. Hippocampal tissue was used to assess levels of NMDAR, IL-1ß, TNF-α, MDA, SOD, NOS, and apoptosis markers. Histological assessment and TUNEL staining were performed in CA1 hippocampal region. RESULTS: The c/tDCS significantly decreased the levels of IL-1ß and TNF-α than the I and a/tDCS groups. The c/tDCS significantly reduced MDA and NOS levels, while increasing the level of SOD than the I and a/tDCS. The c/tDCS caused a significant decrease in NMDAR level than the a/tDCS. Using c/tDCS significantly reduced the Bax and Caspase-3 expressions, while increasing the Bcl-2 expression than the I group. In the c/tDCS group, DNA fragmentation and neuronal death were significantly lower than the I and a/tDCS groups. CONCLUSION: Using cathodal a direct current could attenuate primary pathophysiological pathways induced by CI, and it eventually reduced neurons death and apoptosis in the CA1 hippocampal region.


Assuntos
Isquemia Encefálica , Região CA1 Hipocampal , Estimulação Transcraniana por Corrente Contínua , Animais , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Região CA1 Hipocampal/fisiopatologia , Masculino , Neuroproteção , Ratos , Ratos Wistar , Resultado do Tratamento
5.
Exp Neurol ; 350: 113969, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34973962

RESUMO

Gradual decline in cognitive and non-cognitive functions are considered clinical hallmarks of Alzheimer's Disease (AD). Post-mortem autoptic analysis shows the presence of amyloid ß deposits, neuroinflammation and severe brain atrophy. However, brain circuit alterations and cellular derailments, assessed in very early stages of AD, still remain elusive. The understanding of these early alterations is crucial to tackle defective mechanisms. In a previous study we proved that the Tg2576 mouse model of AD displays functional deficits in the dorsal hippocampus and relevant behavioural AD-related alterations. We had shown that these deficits in Tg2576 mice correlate with the precocious degeneration of dopamine (DA) neurons in the Ventral Tegmental Area (VTA) and can be restored by L-DOPA treatment. Due to the distinct functionality and connectivity of dorsal versus ventral hippocampus, here we investigated neuronal excitability and synaptic functionality in the ventral CA1 hippocampal sub-region of Tg2576 mice. We found an age-dependent alteration of cell excitability and firing in pyramidal neurons starting at 3 months of age, that correlates with reduced levels in the ventral CA1 of tyrosine hydroxylase - the rate-limiting enzyme of DA synthesis. Additionally, at odds with the dorsal hippocampus, we found no alterations in basal glutamatergic transmission and long-term plasticity of ventral neurons in 8-month old Tg2576 mice compared to age-matched controls. Last, we used computational analysis to model the early derailments of firing properties observed and hypothesize that the neuronal alterations found could depend on dysfunctional sodium and potassium conductances, leading to anticipated depolarization-block of action potential firing. The present study depicts that impairment of cell excitability and homeostatic control of firing in ventral CA1 pyramidal neurons is a prodromal feature in Tg2576 AD mice.


Assuntos
Doença de Alzheimer/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Fenômenos Eletrofisiológicos , Células Piramidais , Potenciais de Ação , Envelhecimento , Animais , Dopaminérgicos/farmacologia , Neurônios Dopaminérgicos , Feminino , Levodopa/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Canais de Potássio , Canais de Sódio , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/fisiopatologia
6.
Cell Rep ; 37(10): 110094, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879272

RESUMO

Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.


Assuntos
Comportamento Animal , Região CA1 Hipocampal/metabolismo , Cognição , Encefalomielite Autoimune Experimental/metabolismo , Interleucina-17/metabolismo , Plasticidade Neuronal , Receptores de Interleucina-17/metabolismo , Sinapses/metabolismo , Animais , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Encefalomielite Autoimune Experimental/psicologia , Interleucina-17/genética , Potenciação de Longa Duração , Masculino , Camundongos Biozzi , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-17/genética , Transdução de Sinais , Aprendizagem Espacial , Sinapses/patologia , Proteínas Quinases p38 Ativadas por Mitógeno
7.
Stem Cell Reports ; 16(12): 3005-3019, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34861165

RESUMO

New neurons are abnormal in the adult hippocampus of Alzheimer's disease (AD) mouse models. The effects of modulating adult neurogenesis on AD pathogenesis differ from study to study. We reported recently that ablation of adult neural stem cells (aNSCs) was associated with improved memory in AD models. Here, we found that long-term potentiation (LTP) was improved in the hippocampus of APP/PS1 mice after ablation of aNSCs. This effect was confirmed in hAPP-J20 mice, a second AD mouse model. On the other hand, we found that exposure to enriched environment (EE) dramatically increased the number of DCX+ neurons, promoted dendritic growth, and affected the location of newborn neurons in the dentate gyrus of APP/PS1 mice, and EE exposure significantly ameliorated memory deficits in APP/PS1 mice. Together, our data suggest that both inhibiting abnormal adult neurogenesis and enhancing healthy adult neurogenesis could be beneficial for AD, and they are not mutually exclusive.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/fisiopatologia , Cognição/fisiologia , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Giro Denteado/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Humanos , Potenciação de Longa Duração , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Presenilina-1/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de GABA-A/metabolismo , Memória Espacial
8.
Nat Commun ; 12(1): 6810, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34815409

RESUMO

The prefrontal-hippocampal dysfunction that underlies cognitive deficits in mental disorders emerges during early development. The lateral entorhinal cortex (LEC) is tightly interconnected with both prefrontal cortex (PFC) and hippocampus (HP), yet its contribution to the early dysfunction is fully unknown. Here we show that mice that mimic the dual genetic (G) -environmental (E) etiology (GE mice) of psychiatric risk have poor LEC-dependent recognition memory at pre-juvenile age and abnormal communication within LEC-HP-PFC networks throughout development. These functional and behavioral deficits relate to sparser projections from LEC to CA1 and decreased efficiency of axonal terminals to activate the hippocampal circuits in neonatal GE mice. In contrast, the direct entorhinal drive to PFC is not affected, yet the PFC is indirectly compromised, as target of the under-activated HP. Thus, the entorhinal-hippocampal circuit is already impaired from neonatal age on in GE mice.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Córtex Entorrinal/fisiopatologia , Transtornos Mentais/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Animais , Animais Recém-Nascidos , Disfunção Cognitiva/genética , Disfunção Cognitiva/imunologia , Modelos Animais de Doenças , Feminino , Interação Gene-Ambiente , Humanos , Masculino , Transtornos Mentais/genética , Transtornos Mentais/imunologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Vias Neurais/fisiopatologia , Optogenética , Técnicas de Patch-Clamp , Gravidez
9.
Mol Brain ; 14(1): 147, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556177

RESUMO

Hypoxia typically accompanies acute inflammatory responses in patients and animal models. However, a limited number of studies have examined the effect of hypoxia in combination with inflammation (Hypo-Inf) on neural function. We previously reported that neuronal excitability in hippocampal CA1 neurons decreased during hypoxia and greatly rebounded upon reoxygenation. We attributed this altered excitability mainly to the dynamic regulation of hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels and input resistance. However, the molecular mechanisms underlying input resistance changes by Hypo-Inf and reperfusion remained unclear. In the present study, we found that a change in the density of the delayed rectifier potassium current (IDR) can explain the input resistance variability. Furthermore, voltage-dependent inactivation of A-type potassium (IA) channels shifted in the depolarizing direction during Hypo-Inf and reverted to normal upon reperfusion without a significant alteration in the maximum current density. Our results indicate that changes in the input resistance, and consequently excitability, caused by Hypo-Inf and reperfusion are at least partially regulated by the availability and voltage dependence of KV channels. Moreover, these results suggest that selective KV channel modulators can be used as potential neuroprotective drugs to minimize hypoxia- and reperfusion-induced neuronal damage.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Hipóxia Celular/fisiologia , Canais de Potássio de Retificação Tardia/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Potenciais de Ação/fisiologia , Animais , Meios de Cultura/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/fisiologia , Técnicas In Vitro , Inflamação , Cinética , Potenciais da Membrana/fisiologia , Fármacos Neuroprotetores/farmacologia , Técnicas de Patch-Clamp , Ratos , Reperfusão , Tetrodotoxina/farmacologia
10.
Mol Neurobiol ; 58(11): 5756-5771, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34403042

RESUMO

Aging is an irreversible biological process that involves oxidative stress, neuroinflammation, and apoptosis, and eventually leads to cognitive dysfunction. However, the underlying mechanisms are not fully understood. In this study, we investigated the role and potential mechanisms of Synaptotagmin-7, a calcium membrane transporter in cognitive impairment in aging mice. Our results indicated that Synaptotagmin-7 expression significantly decreased in the hippocampus of D-galactose-induced or naturally aging mice when compared with healthy controls, as detected by western blot and quantitative reverse transcriptase-polymerase chain reaction analysis. Synaptotagmin-7 overexpression in the dorsal CA1 of the hippocampus reversed long-term potentiation and improved hippocampus-dependent spatial learning in D-galactose-induced aging mice. Synaptotagmin-7 overexpression also led to fully preserved learning and memory in 6-month-old mice. Mechanistically, we demonstrated that Synaptotagmin-7 improved learning and memory by elevating the level of fEPSP and downregulating the expression of aging-related genes such as p53 and p16. The results of our study provide new insights into the role of Synaptotagmin-7 in improving neuronal function and overcoming memory impairment caused by aging, suggesting that Synaptotagmin-7 overexpression may be an innovative therapeutic strategy for treating cognitive impairment.


Assuntos
Envelhecimento/psicologia , Região CA1 Hipocampal/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Sinaptotagminas/fisiologia , Envelhecimento/metabolismo , Animais , Transtornos Cognitivos/terapia , Condicionamento Clássico , Dependovirus/genética , Eletrochoque , Medo/fisiologia , Galactose/toxicidade , Regulação da Expressão Gênica , Genes Reporter , Genes p16 , Genes p53 , Vetores Genéticos/administração & dosagem , Potenciação de Longa Duração , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/fisiopatologia , Transtornos da Memória/terapia , Camundongos , Camundongos Endogâmicos C57BL , Teste do Labirinto Aquático de Morris , Distribuição Aleatória , Reconhecimento Psicológico , Proteínas Recombinantes/metabolismo , Aprendizagem Espacial/efeitos dos fármacos , Organismos Livres de Patógenos Específicos , Sinaptotagminas/genética
11.
Neurobiol Dis ; 158: 105454, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34333153

RESUMO

Patients with Alzheimer's disease (AD) often have fragmentation of sleep/wake cycles and disrupted 24-h (circadian) activity. Despite this, little work has investigated the potential underlying day/night disruptions in cognition and neuronal physiology in the hippocampus. The molecular clock, an intrinsic transcription-translation feedback loop that regulates circadian behavior, may also regulate hippocampal neurophysiological activity. We hypothesized that disrupted diurnal variation in clock gene expression in the hippocampus corresponds with loss of normal day/night differences in membrane excitability, synaptic physiology, and cognition. We previously reported disrupted circadian locomotor rhythms and neurophysiological output of the suprachiasmatic nucleus (the primary circadian clock) in Tg-SwDI mice with human amyloid-beta precursor protein mutations. Here, we report that Tg-SwDI mice failed to show day/night differences in a spatial working memory task, unlike wild-type controls that exhibited enhanced spatial working memory at night. Moreover, Tg-SwDI mice had lower levels of Per2, one of the core components of the molecular clock, at both mRNA and protein levels when compared to age-matched controls. Interestingly, we discovered neurophysiological impairments in area CA1 of the Tg-SwDI hippocampus. In controls, spontaneous inhibitory post-synaptic currents (sIPSCs) in pyramidal cells showed greater amplitude and lower inter-event interval during the day than the night. However, the normal day/night differences in sIPSCs were absent (amplitude) or reversed (inter-event interval) in pyramidal cells from Tg-SwDI mice. In control mice, current injection into CA1 pyramidal cells produced more firing during the night than during the day, but no day/night difference in excitability was observed in Tg-SwDI mice. The normal day/night difference in excitability in controls was blocked by GABA receptor inhibition. Together, these results demonstrate that the normal diurnal regulation of inhibitory transmission in the hippocampus is diminished in a mouse model of AD, leading to decreased daytime inhibition onto hippocampal CA1 pyramidal cells. Uncovering disrupted day/night differences in circadian gene regulation, hippocampal physiology, and memory in AD mouse models may provide insight into possible chronotherapeutic strategies to ameliorate Alzheimer's disease symptoms or delay pathological onset.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Ritmo Circadiano/genética , Regulação da Expressão Gênica/genética , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Memória Espacial , Transmissão Sináptica , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Antagonistas GABAérgicos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Piramidais , Receptor PAR-2/biossíntese , Receptor PAR-2/genética
12.
J Cell Mol Med ; 25(14): 7028-7038, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34121317

RESUMO

Findings from recent studies have revealed that microRNAs (miRNAs) are related to numerous neurological disorders. However, whether miRNAs regulate neuronal anomalies involved in the pathogenesis of depression remain unclear. In the present study, we screened miRNA expression profiles in the CA1 hippocampus of a rat model of depression and found that a specific miRNA, microRNA-211-5p, was significantly down-regulated in depressed rats. When miR-211-5p was up-regulated in these rats, neuronal apoptosis within the CA1 area was suppressed, effects which were accompanied with an amelioration of depression-like behaviours in these rats. These neuroprotective effects of miR-211-5p in depressed rats appear to result through suppression of the Dyrk1A/ASK1/JNK signalling pathway within the CA1 area. In further support of this proposal are the findings that knock-down of miR-211-5p within the CA1 area of normal rats activated the Dyrk1A/ASK1/JNK pathway, resulting in the promotion of neuronal apoptosis and display of depression-like behaviours in these rats. Taken together, these results demonstrate that deficits in miR-211-5p contribute to neuronal apoptosis and thus depression-like behaviours in rats. Therefore, the miR-211-5p/Dyrk1A pathway may be critically involved in the pathogenesis of depression and serve as a potential therapeutic target for the treatment of depression.


Assuntos
Apoptose , Depressão/metabolismo , MicroRNAs/metabolismo , Neurônios/metabolismo , Estresse Psicológico/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiopatologia , Depressão/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , Masculino , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Estresse Psicológico/genética
13.
PLoS Comput Biol ; 17(6): e1009115, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34133417

RESUMO

Alzheimer's Disease (AD) is characterized by progressive neurodegeneration and cognitive impairment. Synaptic dysfunction is an established early symptom, which correlates strongly with cognitive decline, and is hypothesised to mediate the diverse neuronal network abnormalities observed in AD. However, how synaptic dysfunction contributes to network pathology and cognitive impairment in AD remains elusive. Here, we present a grid-cell-to-place-cell transformation model of long-term CA1 place cell dynamics to interrogate the effect of synaptic loss on network function and environmental representation. Synapse loss modelled after experimental observations in the APP/PS1 mouse model was found to induce firing rate alterations and place cell abnormalities that have previously been observed in AD mouse models, including enlarged place fields and lower across-session stability of place fields. Our results support the hypothesis that synaptic dysfunction underlies cognitive deficits, and demonstrate how impaired environmental representation may arise in the early stages of AD. We further propose that dysfunction of excitatory and inhibitory inputs to CA1 pyramidal cells may cause distinct impairments in place cell function, namely reduced stability and place map resolution.


Assuntos
Doença de Alzheimer/etiologia , Modelos Neurológicos , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Biologia Computacional , Simulação por Computador , Modelos Animais de Doenças , Células de Grade/patologia , Células de Grade/fisiologia , Humanos , Camundongos , Rede Nervosa/patologia , Rede Nervosa/fisiopatologia , Plasticidade Neuronal/fisiologia , Células de Lugar/patologia , Células de Lugar/fisiologia , Sinapses/patologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia
14.
Behav Brain Res ; 412: 113403, 2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34090940

RESUMO

The developmental period is critical in delineating plastic response to internal and external events. However, neurobehavioural effects of global cerebral ischemia (GCI) in the maturing brain remain largely unknown. This study characterised the effects of GCI experienced at puberty on adulthood (1) hippocampus CA1 neuronal damage, (2) cognitive and emotional impairments, and (3) glucocorticoid receptor (GR) expression. Effects of adolescent exposure to the phenol vanillic acid (VA) on post-ischemic outcomes were also determined. Male Long Evans rats (n = 35) were supplemented for 21 consecutive days (postnatal days 33-53) with VA (91 mg/kg) or nut paste vehicle (control) prior to a 10-min GCI or sham surgery. As adults, rats were tested in the Open Field Test (OFT), Elevated-Plus Maze (EPM), and Barnes Maze (BM). GR expression was determined in the basolateral amygdala (BLA), CA1, and paraventricular nucleus (PVN), and brain injury assessed via CA1 neuronal density. Adolescent GCI exposure induced extensive hippocampal CA1 injury, which was not prevented by VA supplementation. Behaviourally, GCI increased EPM exploration while having no impact on spatial memory. VA intake increased OFT peripheral exploration. Notably, while no delayed changes in CA1 and PVN GR immunoreactivity were noted, both treatments separately increased BLA GR expression when compared with sham-nut paste rats. Age at GCI occurrence plays a critical role on post-ischemic impairments. The observation of minimal functional impairments despite important CA1 neuronal damage supports use of compensatory mechanisms. Our findings also show daily VA supplementation during adolescence to have no protective effects on post-ischemic outcomes, contrasting adult intake.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Ácido Vanílico/farmacologia , Fatores Etários , Animais , Isquemia Encefálica/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Suplementos Nutricionais , Hipocampo/metabolismo , Comportamento Impulsivo/fisiologia , Masculino , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Long-Evans , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/fisiologia , Ácido Vanílico/metabolismo
15.
Brain Res Bull ; 173: 97-107, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34022286

RESUMO

Cardiac arrest (CA) is the leading cause of death in humans. Research has shown that mild therapeutic hypothermia (MTH) can reduce neurological sequelae and mortality after CA. Nevertheless, the mechanism remains unclear. This study aimed to determine whether MTH promotes neurogenesis, attenuates neuronal damage, and inhibits apoptosis of neurons in rats after CA. Sprague-Dawley rats were divided into the normothermia and mild hypothermia groups. The rats in the normothermia and hypothermia groups were exposed to 2 h of normothermia (36-37℃) and hypothermia (32-33℃), respectively, immediately after resuscitation from 5 min of asphyxial CA. Corresponding control groups not subjected to CA were included. On days 1-6, 5-bromodeoxyuridine (BrdU) 100 mg/kg/day was administered intraperitoneally. The animals were euthanized 1 week after CA. Compared with the normothermia group, the hypothermia group showed a significant increase in the number of doublecortin (DCX) immune-positive cells in the subgranular zone of the hippocampus 1 week after CA. Neurogenesis was assessed using double immunofluorescent labeling of BrdU with neuronal-specific nuclear protein (NeuN)/DCX. There was no marked change in the number of newborn mature (BrdU+-NeuN+) neurons, though there was a significant increase in the number of newborn immature (BrdU+-DCX+) neurons in the hypothermia than in the normothermia group 1 week after CA. Neuronal injury and apoptosis in the CA1 region of the hippocampus, assessed using NeuN immunofluorescence and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays, were significantly reduced in the hypothermia group 1 week after CA. Moreover, mild hypothermia increased the expression of cold-shock protein RNA-binding motif protein 3 (RBM3) in the early stage (24 h/48 h) after CA. These results suggested that mild hypothermia promotes generation of neuronal cells, reduces neuronal injury, and inhibits apoptosis of neurons, which may be related to RBM3 expression.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Parada Cardíaca/terapia , Hipotermia Induzida , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Parada Cardíaca/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley
16.
Neurobiol Learn Mem ; 183: 107462, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34015444

RESUMO

Autism spectrum disorder (ASD) is a severe life-long neuropsychiatric disorder. Alterations and imbalance of several neurochemical systems may be involved in ASD pathophysiology, of them, serotonergic neurotransmission dysfunction and deficiency may underlie behavioral abnormalities associated with ASD. However, the functional importance of serotonergic receptors, particularly 5HT7 receptors in ASD pathology remains poorly defined. Serotonin receptor subtype 7 (5-HT7R) plays a direct regulatory role in the development and also for the mature function of the brain, therefore, further studies are necessary to elucidate the role of these receptors in the etiology of autism. To address this issue, we combined here behavioral, electrophysiological methods to further characterize the contribution of 5-HT7Rs in the prenatal valproic acid (VPA) exposure-induced impairment in synaptic plasticity and their impact on the associated behavioral changes. This may help to unravel the underlying cellular mechanisms involved in ASD and can lead to new treatment and/or prevention therapies based on the role of the serotonergic system for autism. Findings revealed that compared to control, autistic-like offspring showed increased anxiety-like behavior, reduced social interaction, decreased locomotor activity, and impaired identification of the novel object. However, administration of 5-HT7Rs agonist, LP-211, for 7 consecutive days before testing from postnatal day 21 to 27 reversed all behavioral deficits induced by prenatal exposure to VPA in offspring. Also, both short-term depression and long-term potentiation were impaired in the autistic-like pups, but activation of 5-HT7Rs rescued the LTP impairment in the autistic-like group so that there was no significant difference between the two groups. Blockade of 5-HT7Rs caused LTP impairment following HFS in the autistic-like group. Besides, there was a significant difference in LTD induction following SB-269970 application between the control and the autistic-like groups measured at first 10 min following TPS. Moreover, both the number and the size of retrograde fast blue-labelled neurons in the raphe nuclei were reduced. Overall, these results provide for the first time, as far as we know, functional evidence for the restorative role of 5-HT7Rs activation against prenatal VPA exposure induced behavioral deficits and hippocampal synaptic plasticity impairment. Therefore, these receptors could be a potential and promising pharmacotherapy target for the treatment of autism.


Assuntos
Transtorno do Espectro Autista/metabolismo , Região CA1 Hipocampal/metabolismo , Potenciação de Longa Duração/fisiologia , Receptores de Serotonina/metabolismo , Animais , Transtorno do Espectro Autista/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiopatologia , Modelos Animais de Doenças , Teste de Labirinto em Cruz Elevado , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , GABAérgicos/toxicidade , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Teste de Campo Aberto , Fenóis/farmacologia , Piperazinas/farmacologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Núcleos da Rafe/metabolismo , Núcleos da Rafe/patologia , Ratos , Receptores de Serotonina/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Comportamento Social , Sulfonamidas/farmacologia , Ácido Valproico/toxicidade
17.
Cell Rep ; 35(3): 109008, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33882308

RESUMO

Synapse loss and altered synaptic strength are thought to underlie cognitive impairment in Alzheimer's disease (AD) by disrupting neural activity essential for memory. While synaptic dysfunction in AD has been well characterized in anesthetized animals and in vitro, it remains unknown how synaptic transmission is altered during behavior. By measuring synaptic efficacy as mice navigate in a virtual reality task, we find deficits in interneuron connection strength onto pyramidal cells in hippocampal CA1 in the 5XFAD mouse model of AD. These inhibitory synaptic deficits are most pronounced during sharp-wave ripples, network oscillations important for memory that require inhibition. Indeed, 5XFAD mice exhibit fewer and shorter sharp-wave ripples with impaired place cell reactivation. By showing inhibitory synaptic dysfunction in 5XFAD mice during spatial navigation behavior and suggesting a synaptic mechanism underlying deficits in network activity essential for memory, this work bridges the gap between synaptic and neural activity deficits in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Interneurônios/metabolismo , Células Piramidais/metabolismo , Navegação Espacial/fisiologia , Sinapses/metabolismo , Doença de Alzheimer/metabolismo , Animais , Ondas Encefálicas/fisiologia , Região CA1 Hipocampal/metabolismo , Modelos Animais de Doenças , Eletrodos Implantados , Humanos , Interneurônios/patologia , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Piramidais/patologia , Sinapses/patologia , Transmissão Sináptica/fisiologia , Realidade Virtual
18.
Behav Brain Res ; 409: 113320, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-33901433

RESUMO

Taste recognition memory is evident in rodents because the initial neophobia to novel tastes attenuates across exposures as the taste becomes familiar and safe. This attenuation of taste neophobia (AN) is context-dependent and an auditory background change could induce the recovery of the neophobic response. The AN auditory context-dependency requires the hippocampal integrity but the neurochemical mechanisms underlying the interaction with the taste memory circuit remain unexplored. We have applied pharmacological intervention by 6-hidroxydopamine (6-OHDA) hippocampal lesion for assessing the role of catecholamines in the hippocampal system to Wistar rats that drank a novel 3% vinegar solution for several consecutive days. Additionally, we manipulated the auditory background as a context that could either change or remain constant across all the drinking sessions. We found that a disruption of the context-dependent AN was induced by intracerebral administration of 6-OHDA targeted to the ventral CA1 hippocampus (vCA1). We conclude that the ability of the auditory context to modulate taste recognition memory involves the catecholaminergic activity in the ventral hippocampal circuit for the proper acquisition of safe taste memory.


Assuntos
Adrenérgicos/farmacologia , Córtex Auditivo/fisiologia , Percepção Auditiva/fisiologia , Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiopatologia , Catecolaminas/metabolismo , Oxidopamina/farmacologia , Reconhecimento Psicológico/fisiologia , Percepção Gustatória/fisiologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Masculino , Ratos Wistar
19.
Int J Mol Sci ; 22(5)2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33807989

RESUMO

For a better translation from treatment designs of schizophrenia to clinical efficiency, there is a crucial need to refine preclinical animal models. In order to consider the multifactorial nature of the disorder, a new mouse model associating three factors (genetic susceptibility-partial deletion of the MAP6 gene, early-life stress-maternal separation, and pharmacological treatment-chronic Δ-9-tetrahydrocannabinol during adolescence) has recently been described. While this model depicts a schizophrenia-like phenotype, the neurobiological correlates remain unknown. Synaptic transmission and functional plasticity of the CA1 hippocampal region of male and female 3-hit mice were therefore investigated using electrophysiological recordings on the hippocampus slice. While basal excitatory transmission remained unaffected, NMDA receptor (NMDAr)-mediated long-term potentiation (LTP) triggered by theta-burst (TBS) but not by high-frequency (HFS) stimulation was impaired in 3-hit mice. Isolated NMDAr activation was not affected or even increased in female 3-hit mice, revealing a sexual dimorphism. Considering that the regulation of LTP is more prone to inhibitory tone if triggered by TBS than by HFS, the weaker potentiation in 3-hit mice suggests a deficiency of intrinsic GABA regulatory mechanisms. Indeed, NMDAr activation was increased by GABAA receptor blockade in wild-type but not in 3-hit mice. This electrophysiological study highlights dysregulations of functional properties and plasticity in hippocampal networks of 3-hit mice, one of the mechanisms suspected to contribute to the pathophysiology of schizophrenia. It also shows differences between males and females, supporting the sexual dimorphism observed in the disorder. Combined with the previously reported study, the present data reinforce the face validity of the 3-hit model that will help to consider new therapeutic strategies for psychosis.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Potenciação de Longa Duração , Esquizofrenia/fisiopatologia , Transmissão Sináptica , Ritmo Teta , Animais , Região CA1 Hipocampal/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Mutantes , Esquizofrenia/genética , Esquizofrenia/patologia
20.
J Neurovirol ; 27(3): 434-443, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33788140

RESUMO

Rabies is a life-threatening viral infection of the brain. Rabies virus (RABV) merely infects excitable cells including neurons provoking drastic behaviors including negative emotional memories. RABV glycoprotein (RVG) plays a critical role in RABV pathogenesis. RVG interacts with various cytoplasmic PDZ (PSD-95/Dlg/ZO-1) containing proteins through its PDZ binding motif (PBM). PTZ domains have crucial role in formation and function of signal transduction. Hippocampus is one of the cerebral regions that contain high load of viral antigens. We examined impact of RVG expression in the dorsal hippocampus on aversive as well as spatial learning and memory performance in rats. Two microliter of the lentiviral vector (~108 T.U./ml) encoding RVG or ∆RVG (deleted PBM) genomes was microinjected into the hippocampal CA1. After 1 week, rat's brain was cross-sectioned and RVG/∆RVG-expressing neuronal cells were confirmed by fluorescent microscopy. Passive avoidance and spatial learning and memory were assessed in rats by Shuttle box and Morris water maze (MWM). In the shuttle box, both RVG and ∆RVG decreased the time spent in the dark compartment compared to control (p < 0.05). In MWM, RVG and ∆RVG did not affect the acquisition of spatial task. In the probe test, RVG-expressing rats spent more time in the target quadrant, and also reached the platform position sooner than control group (p < 0.05). Rats expressing ∆RVG significantly swam farther from the hidden platform than RVG group (p < 0.05). Our data indicate RVG expression in the hippocampus strengthens aversive and spatial learning and memory performance. The boosting effect on spatial but not avoidance memory is mediated through PBM.


Assuntos
Aprendizagem da Esquiva , Região CA1 Hipocampal/fisiopatologia , Glicoproteínas/genética , Aprendizagem em Labirinto , Vírus da Raiva/genética , Memória Espacial , Proteínas Virais/genética , Animais , Região CA1 Hipocampal/metabolismo , Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Injeções Intraventriculares , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/patologia , Vírus da Raiva/química , Vírus da Raiva/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Técnicas Estereotáxicas , Transgenes , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...