Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cereb Cortex ; 30(8): 4306-4324, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32147734

RESUMO

Schizophrenia is associated with cognitive and behavioral dysfunctions thought to reflect imbalances in neurotransmission systems. Recent screenings suggested that lack of (functional) syndapin I (PACSIN1) may be linked to schizophrenia. We therefore studied syndapin I KO mice to address the suggested causal relationship to schizophrenia and to analyze associated molecular, cellular, and neurophysiological defects. Syndapin I knockout (KO) mice developed schizophrenia-related behaviors, such as hyperactivity, reduced anxiety, reduced response to social novelty, and an exaggerated novel object response and exhibited defects in dendritic arborization in the cortex. Neuromorphogenic deficits were also observed for a schizophrenia-associated syndapin I mutant in cultured neurons and coincided with a lack of syndapin I-mediated membrane recruitment of cytoskeletal effectors. Syndapin I KO furthermore caused glutamatergic hypofunctions. Syndapin I regulated both AMPAR and NMDAR availabilities at synapses during basal synaptic activity and during synaptic plasticity-particularly striking were a complete lack of long-term potentiation and defects in long-term depression in syndapin I KO mice. These synaptic plasticity defects coincided with alterations of postsynaptic actin dynamics, synaptic GluA1 clustering, and GluA1 mobility. Both GluA1 and GluA2 were not appropriately internalized. Summarized, syndapin I KO led to schizophrenia-like behavior, and our analyses uncovered associated molecular and cellular mechanisms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Encéfalo/metabolismo , Plasticidade Neuronal/fisiologia , Esquizofrenia/metabolismo , Animais , Comportamento Animal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
2.
Neuroimage ; 188: 347-356, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30553915

RESUMO

Electrical stimulation of right Schaffer collateral in Trpm4-/- knockout and wild type rats were used to study the role of Trpm4 channels for signal processing in the hippocampal formation. Stimulation induced neuronal activity was simultaneously monitored in the CA1 region by in vivo extracellular field recordings and in the entire brain by BOLD fMRI measurements. In wild type and Trpm4-/- knockout rats, consecutive 5 Hz pulse trains elicited similar neuronal responses in the CA1 region and similar BOLD responses in the stimulated right hippocampus. Stimulus-related positive BOLD responses were also found in the left dorsal hippocampus. In contrast to the right dorsal hippocampus, baseline BOLD signals in the left hippocampus significantly decreased during consecutive stimulation trains. Similarly, slowly developing significant declines in baseline BOLD signals, in absence of any positive BOLD responses, were also observed in the right entorhinal, right piriform cortex, right basolateral amygdala and right dorsal striatum whereas baseline BOLD signals remained almost stable in the corresponding left regions. Furthermore, significant declines in baseline BOLD signals were found in the prefrontal cortex and prelimbic/infralimbic cortex. Because significant baseline BOLD declines were only observed in target regions of the right dorsal hippocampus, it might reflect functional connectivity between these regions. In all observed regions the decline in baseline BOLD signals was significantly delayed and less pronounced in Trpm4-/- knockout rats when compared to wild type rats. Thus, either Trpm4 channels are involved in mediating these baseline BOLD shifts or functional connectivity of the hippocampus is impaired in Trpm4-/- knockout rats.


Assuntos
Hipocampo/fisiologia , Canais de Cátion TRPM/fisiologia , Animais , Região CA1 Hipocampal/diagnóstico por imagem , Região CA1 Hipocampal/fisiologia , Estimulação Elétrica , Eletrocorticografia , Feminino , Lateralidade Funcional/fisiologia , Hipocampo/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Transgênicos
3.
Neurobiol Dis ; 125: 14-22, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30665005

RESUMO

Accumulation of hyper-phosphorylated and aggregated Tau proteins is a neuropathological hallmark of Alzheimer's Disease (AD) and Tauopathies. AD patient brains also exhibit insulin resistance. Whereas, under normal physiological conditions insulin signaling in the brain mediates plasticity and memory formation, it can also regulate peripheral energy homeostasis. Thus, in AD, brain insulin resistance affects both cognitive and metabolic changes described in these patients. While a role of Aß oligomers and APOE4 towards the development of brain insulin resistance emerged, contribution of Tau pathology has been largely overlooked. Our recent data demonstrated that one of the physiological function of Tau is to sustain brain insulin signaling. We postulated that under pathological conditions, hyper-phosphorylated/aggregated Tau is likely to lose this function and to favor the development of brain insulin resistance. This hypothesis was substantiated by observations from patient brains with pure Tauopathies. To address the potential link between Tau pathology and brain insulin resistance, we have evaluated the brain response to insulin in a transgenic mouse model of AD-like Tau pathology (THY-Tau22). Using electrophysiological and biochemical evaluations, we surprisingly observed that, at a time when Tau pathology and cognitive deficits are overt and obvious, the hippocampus of THY-Tau22 mice exhibits enhanced response to insulin. In addition, we demonstrated that the ability of i.c.v. insulin to promote body weight loss is enhanced in THY-Tau22 mice. In line with this, THY-Tau22 mice exhibited a lower body weight gain, hypoleptinemia and hypoinsulinemia and finally a metabolic resistance to high-fat diet. The present data highlight that the brain of transgenic Tau mice exhibit enhanced brain response to insulin. Whether these observations are ascribed to the development of Tau pathology, and therefore relevant to human Tauopathies, or unexpectedly results from the Tau transgene overexpression is debatable and discussed.


Assuntos
Encéfalo/metabolismo , Insulina/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas tau/genética
4.
Neurobiol Dis ; 113: 82-96, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29427755

RESUMO

Type 2 diabetes (T2DM) and obesity might increase the risk for AD by 2-fold. Different attempts to model the effect of diet-induced diabetes on AD pathology in transgenic animal models, resulted in opposite conclusions. Here, we used a novel knock-in mouse model for AD, which, differently from other models, does not overexpress any proteins. Long-term high fat diet treatment triggers a reduction in hippocampal N-acetyl-aspartate/myo-inositol metabolites ratio and impairs long term potentiation in hippocampal acute slices. Interestingly, these alterations do not correlate with changes in the core neuropathological features of AD, i.e. amyloidosis and Tau hyperphosphorylation. The data suggest that AD phenotypes associated with high fat diet treatment seen in other models for AD might be exacerbated because of the overexpressing systems used to study the effects of familial AD mutations. Our work supports the increasing insight that knock-in mice might be more relevant models to study the link between metabolic disorders and AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Potenciação de Longa Duração/fisiologia , Doença de Alzheimer/patologia , Animais , Glicemia/metabolismo , Dieta Hiperlipídica/tendências , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos
5.
Nature ; 486(7402): 256-60, 2012 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-22699619

RESUMO

Autism spectrum disorders comprise a range of neurodevelopmental disorders characterized by deficits in social interaction and communication, and by repetitive behaviour. Mutations in synaptic proteins such as neuroligins, neurexins, GKAPs/SAPAPs and ProSAPs/Shanks were identified in patients with autism spectrum disorder, but the causative mechanisms remain largely unknown. ProSAPs/Shanks build large homo- and heteromeric protein complexes at excitatory synapses and organize the complex protein machinery of the postsynaptic density in a laminar fashion. Here we demonstrate that genetic deletion of ProSAP1/Shank2 results in an early, brain-region-specific upregulation of ionotropic glutamate receptors at the synapse and increased levels of ProSAP2/Shank3. Moreover, ProSAP1/Shank2(-/-) mutants exhibit fewer dendritic spines and show reduced basal synaptic transmission, a reduced frequency of miniature excitatory postsynaptic currents and enhanced N-methyl-d-aspartate receptor-mediated excitatory currents at the physiological level. Mutants are extremely hyperactive and display profound autistic-like behavioural alterations including repetitive grooming as well as abnormalities in vocal and social behaviours. By comparing the data on ProSAP1/Shank2(-/-) mutants with ProSAP2/Shank3αß(-/-) mice, we show that different abnormalities in synaptic glutamate receptor expression can cause alterations in social interactions and communication. Accordingly, we propose that appropriate therapies for autism spectrum disorders are to be carefully matched to the underlying synaptopathic phenotype.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Transtorno Autístico/genética , Comportamento Animal/fisiologia , Proteínas do Tecido Nervoso/genética , Agitação Psicomotora/genética , Animais , Transtorno Autístico/patologia , Espinhas Dendríticas/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Agitação Psicomotora/patologia , Receptores Ionotrópicos de Glutamato/metabolismo , Sinapses/metabolismo , Regulação para Cima , Vocalização Animal/fisiologia
6.
Pflugers Arch ; 468(4): 593-607, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26631168

RESUMO

TRPM4 is a calcium-activated but calcium-impermeable non-selective cation (CAN) channel. Previous studies have shown that TRPM4 is an important regulator of Ca(2+)-dependent changes in membrane potential in excitable and non-excitable cell types. However, its physiological significance in neurons of the central nervous system remained unclear. Here, we report that TRPM4 proteins form a CAN channel in CA1 neurons of the hippocampus and we show that TRPM4 is an essential co-activator of N-methyl-D-aspartate (NMDA) receptors (NMDAR) during the induction of long-term potentiation (LTP). Disrupting the Trpm4 gene in mice specifically eliminates NMDAR-dependent LTP, while basal synaptic transmission, short-term plasticity, and NMDAR-dependent long-term depression are unchanged. The induction of LTP in Trpm4 (-/-) neurons was rescued by facilitating NMDA receptor activation or post-synaptic membrane depolarization. Accordingly, we obtained normal LTP in Trpm4 (-/-) neurons in a pairing protocol, where post-synaptic depolarization was applied in parallel to pre-synaptic stimulation. Taken together, our data are consistent with a novel model of LTP induction in CA1 hippocampal neurons, in which TRPM4 is an essential player in a feed-forward loop that generates the post-synaptic membrane depolarization which is necessary to fully activate NMDA receptors during the induction of LTP but which is dispensable for the induction of long-term depression (LTD). These results have important implications for the understanding of the induction process of LTP and the development of nootropic medication.


Assuntos
Região CA1 Hipocampal/metabolismo , Potenciação de Longa Duração , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciais Sinápticos , Canais de Cátion TRPM/metabolismo , Animais , Região CA1 Hipocampal/citologia , Células Cultivadas , Retroalimentação Fisiológica , Camundongos , Neurônios/fisiologia , Canais de Cátion TRPM/genética
7.
Alzheimers Dement ; 12(9): 964-976, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27107518

RESUMO

INTRODUCTION: In Alzheimer's disease (AD), pathologic amyloid-beta (Aß) is synaptotoxic and impairs neuronal function at the microscale, influencing brain networks at the macroscale before Aß deposition. The latter can be detected noninvasively, in vivo, using resting-state functional MRI (rsfMRI), a technique used to assess brain functional connectivity (FC). METHODS: RsfMRI was performed longitudinally in TG2576 and PDAPP mice, starting before Aß deposition to determine the earliest FC changes. Additionally, the role of pathologic Aß on early FC alterations was investigated by treating TG2576 mice with the 3D6 anti-Aß-antibody. RESULTS: Both transgenic models showed hypersynchronized FC before Aß deposition and hyposynchronized FC at later stages. Early anti-Aß treatment in TG2576 mice prevented hypersynchronous FC and the associated synaptic impairments and excitatory/inhibitory disbalances. DISCUSSION: Hypersynchrony of FC may be used as a new noninvasive read out of early AD and can be recovered by anti-Aß treatment, encouraging preventive treatment strategies in familial AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Doença de Alzheimer/diagnóstico por imagem , Animais , Autoanticorpos/farmacologia , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Circulação Cerebrovascular/fisiologia , Sincronização Cortical/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Estudos Longitudinais , Imageamento por Ressonância Magnética , Camundongos Transgênicos , Vias Neurais/diagnóstico por imagem , Vias Neurais/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Oxigênio/sangue , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/fisiopatologia , Placa Amiloide/prevenção & controle , Sintomas Prodrômicos , Descanso
8.
EMBO J ; 30(24): 4955-69, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21926968

RESUMO

Synaptic transmission relies on effective and accurate compensatory endocytosis. F-BAR proteins may serve as membrane curvature sensors and/or inducers and thereby support membrane remodelling processes; yet, their in vivo functions urgently await disclosure. We demonstrate that the F-BAR protein syndapin I is crucial for proper brain function. Syndapin I knockout (KO) mice suffer from seizures, a phenotype consistent with excessive hippocampal network activity. Loss of syndapin I causes defects in presynaptic membrane trafficking processes, which are especially evident under high-capacity retrieval conditions, accumulation of endocytic intermediates, loss of synaptic vesicle (SV) size control, impaired activity-dependent SV retrieval and defective synaptic activity. Detailed molecular analyses demonstrate that syndapin I plays an important role in the recruitment of all dynamin isoforms, central players in vesicle fission reactions, to the membrane. Consistently, syndapin I KO mice share phenotypes with dynamin I KO mice, whereas their seizure phenotype is very reminiscent of fitful mice expressing a mutant dynamin. Thus, syndapin I acts as pivotal membrane anchoring factor for dynamins during regeneration of SVs.


Assuntos
Neurônios/fisiologia , Neuropeptídeos/fisiologia , Fosfoproteínas/fisiologia , Vesículas Sinápticas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Dinaminas/metabolismo , Endocitose , Hipocampo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Neurônios/ultraestrutura , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Retina/fisiologia , Retina/ultraestrutura , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Convulsões/genética , Transmissão Sináptica , Vesículas Sinápticas/genética , Vesículas Sinápticas/ultraestrutura
9.
J Neurosci ; 33(32): 12915-28, 12928a, 2013 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-23926248

RESUMO

The metalloproteinase ADAM10 is of importance for Notch-dependent cortical brain development. The protease is tightly linked with α-secretase activity toward the amyloid precursor protein (APP) substrate. Increasing ADAM10 activity is suggested as a therapy to prevent the production of the neurotoxic amyloid ß (Aß) peptide in Alzheimer's disease. To investigate the function of ADAM10 in postnatal brain, we generated Adam10 conditional knock-out (A10cKO) mice using a CaMKIIα-Cre deleter strain. The lack of ADAM10 protein expression was evident in the brain cortex leading to a reduced generation of sAPPα and increased levels of sAPPß and endogenous Aß peptides. The A10cKO mice are characterized by weight loss and increased mortality after weaning associated with seizures. Behavioral comparison of adult mice revealed that the loss of ADAM10 in the A10cKO mice resulted in decreased neuromotor abilities and reduced learning performance, which were associated with altered in vivo network activities in the hippocampal CA1 region and impaired synaptic function. Histological and ultrastructural analysis of ADAM10-depleted brain revealed astrogliosis, microglia activation, and impaired number and altered morphology of postsynaptic spine structures. A defect in spine morphology was further supported by a reduction of the expression of NMDA receptors subunit 2A and 2B. The reduced shedding of essential postsynaptic cell adhesion proteins such as N-Cadherin, Nectin-1, and APP may explain the postsynaptic defects and the impaired learning, altered network activity, and synaptic plasticity of the A10cKO mice. Our study reveals that ADAM10 is instrumental for synaptic and neuronal network function in the adult murine brain.


Assuntos
Proteínas ADAM/deficiência , Secretases da Proteína Precursora do Amiloide/deficiência , Encéfalo/ultraestrutura , Espinhas Dendríticas/patologia , Epilepsia/genética , Epilepsia/patologia , Deficiências da Aprendizagem/patologia , Proteínas de Membrana/deficiência , Sinapses/patologia , Proteína ADAM10 , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Caderinas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Moléculas de Adesão Celular/metabolismo , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/genética , Gliose/genética , Deficiências da Aprendizagem/genética , Camundongos , Camundongos Transgênicos , Nectinas , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura
10.
Neurobiol Dis ; 51: 144-51, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23153818

RESUMO

Neurobeachin (NBEA), a brain-enriched multidomain scaffolding protein involved in neurotransmitter release and synaptic functioning, has been identified as a candidate gene for autism spectrum disorder (ASD) in four unrelated patients haploinsufficient for NBEA. The aim of this study was to map the behavioral phenotype of Nbea(+/-) mice in order to understand its contribution to the pathogenesis of ASD. ASD-like behavioral variables of Nbea(+/-) mice were related to basal neuronal activity in different brain regions by in situ hybridizations and extracellular field recordings of synaptic plasticity in hippocampal cornu ammonis 1 (CA1) region. Levels of BDNF and phosphorylated cAMP response element-binding protein (CREB) were measured in an attempt to investigate putatively underlying changes in these neuromolecules. Nbea(+/-) mice exhibit several ASD-like features, including changes in self-grooming behavior, social behaviors, conditioned fear responses, and spatial learning and memory, which coincided with enhanced long-term potentiation (LTP) in their CA1 region. The observed alterations in learning and memory and hippocampal LTP are concomitant with decreased expression of the immediate early gene zif268 in dorsomedial striatum and hippocampal CA1 region, increased CREB phosphorylation, and increased hippocampal BDNF expression. These findings indicate that Nbea haploinsufficiency leads to various molecular and cellular changes that affect neuroplasticity and behavioral functions in mice, and could thus underlie the ASD symptomatology in NBEA deficient humans.


Assuntos
Transtorno Autístico/genética , Comportamento Animal/fisiologia , Encéfalo/fisiopatologia , Proteínas de Transporte/genética , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal/genética , Animais , Criança , Feminino , Haploinsuficiência , Humanos , Immunoblotting , Hibridização In Situ , Aprendizagem/fisiologia , Potenciação de Longa Duração/fisiologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Transmissão Sináptica/genética
11.
Brain Behav Immun ; 33: 15-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23747799

RESUMO

We have previously shown that long-term potentiation (LTP) induces hippocampal IL-1ß and IL-6 over-expression, and interfering their signalling either inhibits or supports, respectively, LTP maintenance. Consistently, blockade of endogenous IL-1 or IL-6 restricts or favours hippocampal-dependent memory, effects that were confirmed in genetically manipulated mice. Since cytokines are known for their high degree of mutual crosstalk, here we studied whether a network of cytokines with known neuromodulatory actions is activated during LTP and learning. We found that, besides IL-1ß and IL-6, also IL-1 receptor antagonist (IL-1ra) and IL-18, but not TNFα are over-expressed during LTP maintenance in freely moving rats. The increased expression of these cytokines is causally related to an increase in synaptic strength since it was abrogated when LTP was interfered by blockade of NMDA-glutamate receptors. Likewise, IL-1 and IL-6 were found to be over-expressed in defined regions of the hippocampus during learning a hippocampus-dependent task. However, during learning, changes in IL-18 were restricted to the dorsal hippocampus, and no differences in TNFα and IL1-ra expression were noticed in the hippocampus. Noticeably, IL-1ra transcripts were significantly reduced in the prefrontal cortex. The relation between cytokine expression and learning was causal because such changes were not observed in animals from a pseudo-trained group that was subject to the same manipulation but could not learn the task. Taken together with previous studies, we conclude that activation of a cytokine network in the brain is a physiologic relevant phenomenon not only for LTP maintenance but also for certain types of learning.


Assuntos
Citocinas/fisiologia , Giro Denteado/imunologia , Aprendizagem/fisiologia , Potenciação de Longa Duração/imunologia , Animais , Células Cultivadas , Células HEK293 , Hipocampo/imunologia , Hipocampo/metabolismo , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/fisiologia , Interleucina-18/fisiologia , Interleucina-1beta/fisiologia , Interleucina-6/fisiologia , Camundongos , Camundongos Mutantes , Receptores Nucleares Órfãos/antagonistas & inibidores , Receptores Nucleares Órfãos/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
12.
Neural Comput ; 25(3): 650-70, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23272921

RESUMO

Recent advances have started to uncover the underlying mechanisms of metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD). However, it is not completely clear how these mechanisms are linked, and it is believed that several crucial mechanisms remain to be revealed. In this study, we investigated whether system identification (SI) methods can be used to gain insight into the mechanisms of synaptic plasticity. SI methods have been shown to be an objective and powerful approach for describing how sensory neurons encode information about stimuli. However, to our knowledge, it is the first time that SI methods have been applied to electrophysiological brain slice recordings of synaptic plasticity responses. The results indicate that the SI approach is a valuable tool for reverse-engineering of mGluR-LTD responses. We suggest that such SI methods can aid in unraveling the complexities of synaptic function.


Assuntos
Algoritmos , Depressão Sináptica de Longo Prazo/fisiologia , Modelos Neurológicos , Receptores de Glutamato Metabotrópico/fisiologia , Animais , Hipocampo/fisiologia , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar
13.
Adv Redox Res ; 7: None, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38798747

RESUMO

Ionising radiation (IR) is a cause of lipid peroxidation, and epidemiological data have revealed a correlation between exposure to IR and the development of eye lens cataracts. Cataracts remain the leading cause of blindness around the world. The plasma membranes of lens fibre cells are one of the most cholesterolrich membranes in the human body, forming lipid rafts and contributing to the biophysical properties of lens fibre plasma membrane. Liquid chromatography followed by mass spectrometry was used to analyse bovine eye lens lipid membrane fractions after exposure to 5 and 50 Gy and eye lenses taken from wholebody 2 Gy-irradiated mice. Although cholesterol levels do not change significantly, IR dose-dependant formation of the oxysterols 7ß-hydroxycholesterol, 7-ketocholesterol and 5, 6-epoxycholesterol in bovine lens nucleus membrane extracts was observed. Whole-body X-ray exposure (2 Gy) of 12-week old mice resulted in an increase in 7ß-hydroxycholesterol and 7-ketocholesterol in their eye lenses. Their increase regressed over 24 h in the living lens cortex after IR exposure. This study also demonstrated that the IR-induced fold increase in oxysterols was greater in the mouse lens cortex than the nucleus. Further work is required to elucidate the mechanistic link(s) between oxysterols and IR-induced cataract, but these data evidence for the first time that IR exposure of mice results in oxysterol formation in their eye lenses.

14.
Alzheimers Res Ther ; 15(1): 16, 2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36641439

RESUMO

BACKGROUND: Hyperphosphorylation and intraneuronal aggregation of the microtubule-associated protein tau is a major pathological hallmark of Alzheimer's disease (AD) brain. Of special interest is the effect of cerebral amyloid beta deposition, the second main hallmark of AD, on human tau pathology. Therefore, studying the influence of cerebral amyloidosis on human tau in a novel human tau knock-in (htau-KI) mouse model could help to reveal new details on their interplay. METHODS: We studied the effects of a novel human htau-KI under fast-progressing amyloidosis in 5xFAD mice in terms of correlation of gene expression data with human brain regions, development of Alzheimer's-like pathology, synaptic transmission, and behavior. RESULTS: The main findings are an interaction of human beta-amyloid and human tau in crossbred 5xFADxhtau-KI observed at transcriptional level and corroborated by electrophysiology and histopathology. The comparison of gene expression data of the 5xFADxhtau-KI mouse model to 5xFAD, control mice and to human AD patients revealed conspicuous changes in pathways related to mitochondria biology, extracellular matrix, and immune function. These changes were accompanied by plaque-associated MC1-positive pathological tau that required the htau-KI background. LTP deficits were noted in 5xFAD and htau-KI mice in contrast to signs of rescue in 5xFADxhtau-KI mice. Increased frequencies of miniature EPSCs and miniature IPSCs indicated an upregulated presynaptic function in 5xFADxhtau-KI. CONCLUSION: In summary, the multiple interactions observed between knocked-in human tau and the 5xFAD-driven progressing amyloidosis have important implications for future model development in AD.


Assuntos
Doença de Alzheimer , Amiloidose , Camundongos , Humanos , Animais , Peptídeos beta-Amiloides/metabolismo , Camundongos Transgênicos , Doença de Alzheimer/patologia , Proteínas tau/genética , Proteínas tau/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo
15.
J Neurosci ; 31(7): 2511-25, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21325519

RESUMO

This report describes the behavioral and electrophysiological analysis of regulatable transgenic mice expressing mutant repeat domains of human Tau (Tau(RD)). Mice were generated to express Tau(RD) in two forms, differing in their propensity for ß-structure and thus in their tendency for aggregation ("pro-aggregant" or "anti-aggregant") (Mocanu et al., 2008). Only pro-aggregant mice show pronounced changes typical for Tau pathology in Alzheimer's disease (aggregation, missorting, hyperphosphorylation, synaptic and neuronal loss), indicating that the ß-propensity and hence the ability to aggregate is a key factor in the disease. We now tested the mice with regard to neuromotor parameters, behavior, learning and memory, and synaptic plasticity and correlated this with histological and biochemical parameters in different stages of switching Tau(RD) on or off. The mice are normal in neuromotor tests. However, pro-aggregant Tau(RD) mice are strongly impaired in memory and show pronounced loss of long-term potentiation (LTP), suggesting that Tau aggregation specifically perturbs these brain functions. Remarkably, when the expression of human pro-aggregant Tau(RD) is switched on for ∼ 10 months and off for ∼ 4 months, memory and LTP recover, whereas aggregates decrease moderately and change their composition from mixed human plus mouse Tau to mouse Tau only. Neuronal loss persists, but synapses are partially rescued. This argues that continuous presence of amyloidogenic pro-aggregant Tau(RD) constitutes the main toxic insult for memory and LTP, rather than the aggregates as such.


Assuntos
Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Mutação/genética , Tauopatias , Proteínas tau/genética , Animais , Modelos Animais de Doenças , Doxiciclina/farmacologia , Força da Mão/fisiologia , Hipocampo/patologia , Hipocampo/ultraestrutura , Humanos , Técnicas In Vitro , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Testes Neuropsicológicos , Estrutura Terciária de Proteína , Percepção Espacial/fisiologia , Sinapses/patologia , Sinapses/ultraestrutura , Tauopatias/metabolismo , Tauopatias/patologia , Tauopatias/fisiopatologia , Fatores de Tempo , Proteínas tau/metabolismo
16.
Acta Neuropathol ; 123(6): 787-805, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22532069

RESUMO

Neurofibrillary lesions of abnormal Tau are hallmarks of Alzheimer disease and frontotemporal dementias. Our regulatable (Tet-OFF) mouse models of tauopathy express variants of human full-length Tau in the forebrain (CaMKIIα promoter) either with mutation ΔK280 (pro-aggregant) or ΔK280/I277P/I308P (anti-aggregant). Co-expression of luciferase enables in vivo quantification of gene expression by bioluminescence imaging. Pro-aggregant mice develop synapse loss and Tau-pathology including missorting, phosphorylation and early pretangle formation, whereas anti-aggregant mice do not. We correlated hippocampal Tau pathology with learning/memory performance and synaptic plasticity. Pro-aggregant mice at 16 months of gene expression exhibited severe cognitive deficits in Morris water maze and in passive-avoidance paradigms, whereas anti-aggregant mice were comparable to controls. Cognitive impairment of pro-aggregant mice was accompanied by loss of hippocampal LTP in CA1 and CA3 areas and by a reduction of synaptic proteins and dendritic spines, although no neuronal loss was observed. Remarkably, memory and LTP recovered when pro-aggregant Tau was switched-OFF for ~4 months, Tau phosphorylation and missorting were reversed, and synapses recovered. Moreover, soluble and insoluble pro-aggregant hTau40 disappeared, while insoluble mouse Tau was still present. This study links early Tau pathology without neurofibrillary tangles and neuronal death to cognitive decline and synaptic dysfunction. It demonstrates that Tau-induced impairments are reversible after switching-OFF pro-aggregant Tau. Therefore, our mouse model may mimic an early phase of AD when the hippocampus does not yet suffer from irreversible cell death but cognitive deficits are already striking. It offers potential to evaluate drugs with regard to learning and memory performance.


Assuntos
Transtornos Cognitivos/metabolismo , Hipocampo/patologia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Transtornos Cognitivos/genética , Modelos Animais de Doenças , Expressão Gênica , Hipocampo/fisiopatologia , Humanos , Aprendizagem/fisiologia , Potenciação de Longa Duração , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Testes Neuropsicológicos , Sinapses/genética , Sinapses/patologia , Tauopatias/metabolismo , Tauopatias/patologia , Fatores de Tempo , Proteínas tau/genética
17.
Neurobiol Learn Mem ; 95(3): 296-304, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21167950

RESUMO

We evaluated various forms of hippocampus-dependent learning and memory, and hippocampal synaptic plasticity in THY-Tau22 transgenic mice, a murine tauopathy model that expresses double-mutated 4-repeat human tau, and shows neuropathological tau hyperphosphorylation and aggregation throughout the brain. Focussing on hippocampus, immunohistochemical studies in aged THY-Tau22 mice revealed prominent hyper- and abnormal phosphorylation of tau in CA1 region, and an increase in glial fibrillary acidic protein (GFAP) in hippocampus, but without signs of neuronal loss. These mice displayed spatial, social, and contextual learning and memory defects that could not be reduced to subtle neuromotor disability. The behavioral defects coincided with changes in hippocampal synaptic functioning and plasticity as measured in paired-pulse and novel long-term depression protocols. These results indicate that hippocampal tauopathy without neuronal cell loss can impair neural and behavioral plasticity, and further show that transgenic mice, such as the THY-Tau22 strain, might be useful for preclinical research on tauopathy pathogenesis and possible treatment.


Assuntos
Aprendizagem por Associação/fisiologia , Hipocampo/fisiopatologia , Depressão Sináptica de Longo Prazo/fisiologia , Aprendizagem em Labirinto/fisiologia , Tauopatias/fisiopatologia , Análise de Variância , Animais , Condicionamento Clássico/fisiologia , Modelos Animais de Doenças , Medo , Preferências Alimentares , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Masculino , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Comportamento Social , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/genética , Proteínas tau/metabolismo
18.
Cereb Cortex ; 20(3): 684-93, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19574394

RESUMO

Vesicular glutamate transporters 1 and 2 (VGLUT1, VGLUT2) show largely complementary distribution in the mature rodent brain and tend to segregate to synapses with different physiological properties. In the hippocampus, VGLUT1 is the dominate subtype in adult animals, whereas VGLUT2 is transiently expressed during early postnatal development. We generated and characterized VGLUT1 knockout mice in order to examine the functional contribution of this transporter to hippocampal synaptic plasticity and hippocampus-dependent spatial learning. Because complete deletion of VGLUT1 resulted in postnatal lethality, we used heterozygous animals for analysis. Here, we report that deletion of VGLUT1 resulted in impaired hippocampal long-term potentiation (LTP) in the CA1 region in vitro. In contrast, heterozygous VGLUT2 mice that were investigated for comparison did not show any changes in LTP. The reduced ability of VGLUT1-deficient mice to express LTP was accompanied by a specific deficit in spatial reversal learning in the water maze. Our data suggest a functional role of VGLUT1 in forms of hippocampal synaptic plasticity that are required to adapt and modify acquired spatial maps to external stimuli and changes.


Assuntos
Aprendizagem da Esquiva/fisiologia , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Comportamento Espacial/fisiologia , Proteína Vesicular 1 de Transporte de Glutamato/fisiologia , Análise de Variância , Animais , Biofísica , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Estimulação Elétrica/métodos , Hipocampo/citologia , Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Estatísticas não Paramétricas , Proteína Vesicular 1 de Transporte de Glutamato/deficiência , Proteína Vesicular 2 de Transporte de Glutamato/deficiência
19.
J Physiol ; 588(Pt 12): 2133-45, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20421286

RESUMO

Bassoon, a protein highly concentrated at the synaptic active zone, is thought to participate in the organization of the cytomatrix at the site of neurotransmitter release. Bassoon is amongst the first proteins to accumulate at newly formed synaptic junctions, raising the question of the functional role of this protein in the early stages of synaptic development. Here we show that the course of synaptic maturation of hippocampal mossy fibre (MF) synapses (glutamatergic synapses with multiple release sites) is markedly altered during the first 2 weeks of postnatal development in mutant mice lacking the central region of Bassoon (Bsn(-/-) mice). At postnatal day 7 (P7), Bsn(-/-) mice display large amplitude MF-EPSCs with decreased paired pulse ratios, an abnormality which may be linked to deficits in the organization of the presynaptic active zone. Surprisingly, 1 week later, decreased MF-EPSCs amplitude is observed in Bsn(-/-) mice, consistent with the inactivation of a subset of synaptic release sites. Finally, at more mature states a decreased posttetanic potentiation is observed at MF-synapses. These results support the notion that Bassoon is important for organizing the presynaptic active zone during the postnatal maturation of glutamatergic synapses.


Assuntos
Região CA3 Hipocampal/metabolismo , Ácido Glutâmico/metabolismo , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Piramidais/metabolismo , Sinapses/metabolismo , Transmissão Sináptica , Fatores Etários , Animais , Região CA3 Hipocampal/crescimento & desenvolvimento , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Fibras Musgosas Hipocampais/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Plasticidade Neuronal , Terminações Pré-Sinápticas/metabolismo , Potenciais Sinápticos
20.
BMC Neurosci ; 11: 2, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20051123

RESUMO

BACKGROUND: Survivin is a unique member of the inhibitor of apoptosis protein (IAP) family in that it exhibits antiapoptotic properties and also promotes the cell cycle and mediates mitosis as a chromosome passenger protein. Survivin is highly expressed in neural precursor cells in the brain, yet its function there has not been elucidated. RESULTS: To examine the role of neural precursor cell survivin, we first showed that survivin is normally expressed in periventricular neurogenic regions in the embryo, becoming restricted postnatally to proliferating and migrating NPCs in the key neurogenic sites, the subventricular zone (SVZ) and the subgranular zone (SGZ). We then used a conditional gene inactivation strategy to delete the survivin gene prenatally in those neurogenic regions. Lack of embryonic NPC survivin results in viable, fertile mice (SurvivinCamcre) with reduced numbers of SVZ NPCs, absent rostral migratory stream, and olfactory bulb hypoplasia. The phenotype can be partially rescued, as intracerebroventricular gene delivery of survivin during embryonic development increases olfactory bulb neurogenesis, detected postnatally. SurvivinCamcre brains have fewer cortical inhibitory interneurons, contributing to enhanced sensitivity to seizures, and profound deficits in memory and learning. CONCLUSIONS: The findings highlight the critical role that survivin plays during neural development, deficiencies of which dramatically impact on postnatal neural function.


Assuntos
Encéfalo/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Proteínas Associadas aos Microtúbulos/metabolismo , Neurogênese/fisiologia , Convulsões/fisiopatologia , Células-Tronco/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Transtornos Cognitivos/patologia , Inativação Gênica , Proteínas Inibidoras de Apoptose , Interneurônios/patologia , Interneurônios/fisiologia , Deficiências da Aprendizagem/patologia , Deficiências da Aprendizagem/fisiopatologia , Masculino , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Inibição Neural/fisiologia , Neurônios/patologia , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Proteínas Repressoras , Convulsões/patologia , Nicho de Células-Tronco/crescimento & desenvolvimento , Nicho de Células-Tronco/patologia , Nicho de Células-Tronco/fisiopatologia , Células-Tronco/patologia , Survivina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA