Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Biol ; 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37682518

RESUMO

Synaptic vesicles can undergo several modes of exocytosis, endocytosis, and trafficking within individual synapses, and their fates may be linked to differences in the vesicular protein composition. Here, we mapped the intrasynaptic distribution of the synaptic vesicle proteins SV2B and SV2A in glutamatergic synapses of the hippocampus using three-dimensional electron microscopy. SV2B is almost completely absent from both docked vesicles and a distinct cluster of vesicles found near the active zone. In contrast, SV2A was found in all domains of the synapse and was slightly enriched near the active zone. SV2B and SV2A were found on the membrane in the peri-active zone, suggesting recycling from both clusters of vesicles. SV2B knockout mice displayed an increased seizure induction threshold only in a model employing high-frequency stimulation. Our data show that glutamatergic synapses generate molecularly distinct populations of synaptic vesicles and are able to maintain them at steep spatial gradients. The almost complete absence of SV2B from vesicles at the active zone of wildtype mice may explain why SV2A has been found to be more important for vesicle release.

2.
Front Mol Neurosci ; 16: 1324922, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38283700

RESUMO

Activity induced transcription factor ΔFosB plays a key role in different CNS disorders including epilepsy, Alzheimer's disease, and addiction. Recent findings suggest that ΔFosB drives cognitive deficits in epilepsy and together with the emergence of small molecule inhibitors of ΔFosB activity makes it an interesting therapeutic target. However, whether ΔFosB contributes to pathophysiology or provides protection in drug-resistant epilepsy is still unclear. In this study, ΔFosB was specifically downregulated by delivering AAV-shRNA into the hippocampus of chronically epileptic mice using the drug-resistant pilocarpine model of mesial temporal epilepsy (mTLE). Immunohistochemistry analyses showed that prolonged downregulation of ΔFosB led to exacerbation of neuroinflammatory markers of astrogliosis and microgliosis, loss of mossy fibers, and hippocampal granule cell dispersion. Furthermore, prolonged inhibition of ΔFosB using a ΔJunD construct to block ΔFosB signaling in a mouse model of Alzheimer's disease, that exhibits spontaneous recurrent seizures, led to similar findings, with increased neuroinflammation and decreased NPY expression in mossy fibers. Together, these data suggest that seizure-induced ΔFosB, regardless of seizure-etiology, is part of a homeostatic mechanism that protects the epileptic brain from further deterioration.

3.
Nat Commun ; 13(1): 4444, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35915085

RESUMO

During the early stages of Alzheimer's disease (AD) in both mouse models and human patients, soluble forms of Amyloid-ß 1-42 oligomers (Aß42o) trigger loss of excitatory synapses (synaptotoxicity) in cortical and hippocampal pyramidal neurons (PNs) prior to the formation of insoluble amyloid plaques. In a transgenic AD mouse model, we observed a spatially restricted structural remodeling of mitochondria in the apical tufts of CA1 PNs dendrites corresponding to the dendritic domain where the earliest synaptic loss is detected in vivo. We also observed AMPK over-activation as well as increased fragmentation and loss of mitochondrial biomass in Ngn2-induced neurons derived from a new APPSwe/Swe knockin human ES cell line. We demonstrate that Aß42o-dependent over-activation of the CAMKK2-AMPK kinase dyad mediates synaptic loss through coordinated phosphorylation of MFF-dependent mitochondrial fission and ULK2-dependent mitophagy. Our results uncover a unifying stress-response pathway causally linking Aß42o-dependent structural remodeling of dendritic mitochondria to synaptic loss.


Assuntos
Doença de Alzheimer , Mitofagia , Proteínas Quinases Ativadas por AMP/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Dinâmica Mitocondrial , Fragmentos de Peptídeos , Sinapses/metabolismo
4.
Neurobiol Aging ; 107: 142-152, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34433125

RESUMO

With the emergence of disease-modifying therapies for Parkinson's disease, reliable longitudinal markers are needed to quantify pathology and demonstrate disease progression. We developed the A53T-AAV rat model of synucleinopathy by combining longitudinal measures over 12 weeks. We first characterized the progression of the motor and dopaminergic deficits. Then, we monitored the disease progression using the [18F]FMT Positron Emission Tomography (PET) radiotracer. The nigral injection of A53T-AAV led to an increase in phosphorylated α-synuclein on S129, a progressive accumulation of α-synuclein aggregates, and a decrease of dopaminergic function associated with a deterioration of motor activity. The longitudinal monitoring of A53T-AAV rats with [18F]FMT PET showed a progressive reduction of the Kc outcome parameter in the caudate putamen from the lesioned side. Interestingly, the progressive reduction in the [18F]FMT PET signal correlated with defects in the stepping test. In conclusion, we established a progressive rat model of α-synuclein pathology which monitors the deficit longitudinally using both the [18F]FMT PET tracer and behavioral parameters, 2 features that have strong relevance for translational approaches.


Assuntos
Dependovirus , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/fisiologia , Atividade Motora , Doença de Parkinson/diagnóstico por imagem , Doença de Parkinson/fisiopatologia , Sinucleinopatias/diagnóstico por imagem , Sinucleinopatias/fisiopatologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Radioisótopos de Flúor , Masculino , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fosforilação , Tomografia por Emissão de Pósitrons , Agregados Proteicos , Ratos Sprague-Dawley , Sinucleinopatias/metabolismo , Sinucleinopatias/patologia , Tirosina , alfa-Sinucleína/metabolismo
5.
Int J Mol Sci ; 21(19)2020 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-32977526

RESUMO

Seizures are common in patients with high-grade gliomas (30-60%) and approximately 15-30% of glioblastoma (GB) patients develop drug-resistant epilepsy. Reliable animal models are needed to develop adequate treatments for glioma-related epilepsy. Therefore, fifteen rats were inoculated with F98 GB cells (GB group) and four rats with vehicle only (control group) in the right entorhinal cortex. MRI was performed to visualize tumor presence. A subset of seven GB and two control rats were implanted with recording electrodes to determine the occurrence of epileptic seizures with video-EEG recording over multiple days. In a subset of rats, tumor size and expression of tumor markers were investigated with histology or mRNA in situ hybridization. Tumors were visible on MRI six days post-inoculation. Time-dependent changes in tumor morphology and size were visible on MRI. Epileptic seizures were detected in all GB rats monitored with video-EEG. Twenty-one days after inoculation, rats were euthanized based on signs of discomfort and pain. This study describes, for the first time, reproducible tumor growth and spontaneous seizures upon inoculation of F98 cells in the rat entorhinal cortex. The development of this new model of GB-related epilepsy may be valuable to design new therapies against tumor growth and associated epileptic seizures.


Assuntos
Neoplasias Encefálicas , Eletroencefalografia , Epilepsia , Glioma , Neoplasias Experimentais , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Epilepsia/metabolismo , Epilepsia/patologia , Epilepsia/fisiopatologia , Glioma/metabolismo , Glioma/patologia , Glioma/fisiopatologia , Masculino , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/fisiopatologia , Ratos , Ratos Endogâmicos F344
6.
Epilepsia ; 60(7): 1412-1423, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31179549

RESUMO

OBJECTIVE: The cystine/glutamate antiporter system xc- could represent a new target for antiepileptogenic treatments due to its crucial roles in glutamate homeostasis and neuroinflammation. To demonstrate this, we compared epilepsy development and seizure susceptibility in xCT knockout mice (xCT-/- ) and in littermate controls (xCT+/+ ) in different chronic models of epilepsy. METHODS: Mice were surgically implanted with electrodes in the basolateral amygdala and chronically stimulated to develop self-sustained status epilepticus (SSSE); continuous video-electroencephalography monitoring was performed for 28 days after SE and hippocampal histopathology was assessed. Corneal kindling was induced by twice daily electrical stimulation at 6 Hz and maintenance of the fully kindled state was evaluated. Next, messenger RNA (mRNA) and protein levels of xCT and of the proteins involved in the phosphoinositide 3-kinase (PI3K)/Akt/glycogen synthase kinase 3ß (GSK-3ß)/eukaryotic initiation factor 2α (eIF2α)/activating transcription factor 4 (ATF4) signaling pathway were measured at different time points during epileptogenesis in NMRI mice treated with pilocarpine. Finally, the anticonvulsant effect of sulfasalazine (SAS), a nonselective system xc- inhibitor, was assessed against 6 Hz-evoked seizures in pilocarpine-treated mice. RESULTS: In the SSSE model, xCT-/- mice displayed a significant delayed epileptogenesis, a reduced number of spontaneous recurrent seizures, and less pronounced astrocytic and microglial activation. Moreover, xCT-/- mice showed reduced seizure severity during 6 Hz kindling development and a lower incidence of generalized seizures during the maintenance of the fully kindled state. In pilocarpine-treated mice, protein levels of the PI3K/Akt/GSK-3ß/eIF2α/ATF4 pathway were increased during the chronic phase of the model, consistent with previous findings in the hippocampus of patients with epilepsy. Finally, repeated administration of SAS protected pilocarpine-treated mice against acute 6 Hz seizure induction, in contrast to sham controls, in which system xc- is not activated. SIGNIFICANCE: Inhibition of system xc- could be an attractive target for the development of new therapies with a potential for disease modification in epilepsy.


Assuntos
Sistema y+ de Transporte de Aminoácidos/efeitos dos fármacos , Anticonvulsivantes/farmacologia , Epilepsia/tratamento farmacológico , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Anticonvulsivantes/uso terapêutico , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/etiologia , Epilepsia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pilocarpina/farmacologia , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/etiologia , Estado Epiléptico/metabolismo
7.
Brain ; 142(6): 1736-1750, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31038156

RESUMO

Tauopathies are neurodegenerative diseases characterized by the intraneuronal accumulation of aggregated tau. The staging of this neurodegenerative process is well established for Alzheimer's disease as well as for other tauopathies. The stereotypical pattern of tau pathology in these diseases is consistent with the hypothesis that the tau protein can spread in a 'prion-like' manner. It proposes that extracellular pathological tau species can transmit pathology from cell to cell. Accordingly, by targeting these spreading species with therapeutic antibodies one should be able to slow or halt the progression of tau pathology. To be effective, antibodies should neutralize the pathological species present in Alzheimer's disease brains and block their cell-to-cell spread. To evaluate both aspects, tau antibody D, which recognizes an epitope in the central region of tau, and was selected for its outstanding ability to block tau seeding in cell based assays, was used in this study. Here, we addressed two fundamental questions: (i) can this anti-tau antibody neutralize the pathological species present in Alzheimer's disease brains; and (ii) can it block the cell-to-cell spread of tau seeds in vivo? First, antibody D effectively prevented the induction of tau pathology in the brains of transgenic mice that had been injected with human Alzheimer's disease brain extracts, showing that it could effectively neutralize the pathological species present in these extracts. Second, by using K18 P301L tau fibrils to induce pathology, we further demonstrated that antibody D was also capable of blocking the progression of tau pathology to distal brain regions. In contrast, an amino-terminal tau antibody, which was less effective at blocking tau seeding in vitro showed less efficacy in reducing Alzheimer's disease patient tau driven pathology in the transgenic mouse model. We did not address whether the same is true for a spectrum of other amino-terminal antibodies that were tested in vitro. These data highlight important differences between tau antibodies and, when taken together with other recently published data, suggest that epitope may be important for function.


Assuntos
Doença de Alzheimer/patologia , Emaranhados Neurofibrilares/patologia , Tauopatias/metabolismo , Proteínas tau/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Animais , Anticorpos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Progressão da Doença , Epitopos , Feminino , Fatores Imunológicos/metabolismo , Imunoterapia , Masculino , Camundongos Transgênicos , Proteínas tau/metabolismo
8.
Acta Neuropathol ; 136(5): 729-745, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30238240

RESUMO

In Alzheimer's disease (AD) and other tauopathies, the cytosolic protein Tau misfolds and forms intracellular aggregates which accumulate within the brain leading to neurodegeneration. Clinical progression is tightly linked to the progressive spread of Tau pathology throughout the brain, and several lines of evidence suggest that Tau aggregates or "seeds" may propagate pathology by spreading from cell to cell in a "prion like" manner. Accordingly, blocking the spread of extracellular seeds with an antibody could be a viable therapeutic approach. However, as the structure of Tau seeds is unknown, it is only possible to rationally design therapeutic Tau antibodies by making a priori assumptions. To avoid this, we developed a robust and quantitative cell based assay and employed an unbiased screening approach to identify the antibody with the highest activity against human Tau seeds. The selected antibody (D), directed to the mid-region of Tau (amino acids 235-250), potently blocked the seeding of human AD Tau and was also fully efficacious against seeds from progressive supranuclear palsy. When we compared this antibody with previously described reference antibodies, we were surprised to find that none of these antibodies showed comparable efficacy against human pathological seeds. Our data highlight the difficulty of predicting antibody accessible epitopes on pathological Tau seeds and question the potential efficacy of some of the Tau antibodies that are currently in clinical development.


Assuntos
Anticorpos/metabolismo , Epitopos/imunologia , Proteínas tau/química , Proteínas tau/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Mapeamento de Epitopos , Epitopos/química , Células HEK293 , Humanos , Agregados Proteicos , Conformação Proteica , Ressonância de Plasmônio de Superfície , Transfecção , Proteínas tau/genética , Proteínas tau/metabolismo
9.
Nat Commun ; 9(1): 3561, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30177815

RESUMO

The identification of drug targets is highly challenging, particularly for diseases of the brain. To address this problem, we developed and experimentally validated a general computational framework for drug target discovery that combines gene regulatory information with causal reasoning ("Causal Reasoning Analytical Framework for Target discovery"-CRAFT). Using a systems genetics approach and starting from gene expression data from the target tissue, CRAFT provides a predictive framework for identifying cell membrane receptors with a direction-specified influence over disease-related gene expression profiles. As proof of concept, we applied CRAFT to epilepsy and predicted the tyrosine kinase receptor Csf1R as a potential therapeutic target. The predicted effect of Csf1R blockade in attenuating epilepsy seizures was validated in three pre-clinical models of epilepsy. These results highlight CRAFT as a systems-level framework for target discovery and suggest Csf1R blockade as a novel therapeutic strategy in epilepsy. CRAFT is applicable to disease settings other than epilepsy.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia do Lobo Temporal/genética , Epilepsia/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Animais , Simulação por Computador , Modelos Animais de Doenças , Descoberta de Drogas , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Terapia de Alvo Molecular , Agonistas Muscarínicos/toxicidade , Pilocarpina/toxicidade , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Análise de Sequência de RNA , Biologia de Sistemas
10.
Mov Disord ; 33(2): 273-281, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29278274

RESUMO

BACKGROUND: Lilly/Avid's AV-1451 is one of the most advanced tau PET tracers in the clinic. Although results obtained in Alzheimer's disease patients are compelling, discrimination of tracer uptake in healthy individuals and patients with supranuclear palsy (PSP) is less clear as there is substantial overlap of signal in multiple brain regions. Moreover, accurate quantification of [18 F]AV-1451 uptake in Alzheimer's disease may not be possible. OBJECTIVES: The aim of the present study was to characterize the in vitro binding of AV-1451 to understand and identify potential off-target binding that could explain the poor discrimination observed in PSP patients. METHODS: [3 H]AV-1451 and AV-1451 were characterized in in vitro binding assays using recombinant and native proteins/tissues from postmortem samples of controls and Alzheimer's disease and PSP patients. RESULTS: [3 H]AV-1451 binds to multiple sites with nanomolar affinities in brain homogenates and to tau fibrils isolated from Alzheimer's disease or PSP patients. [3 H]AV-1451 also binds with similarly high affinities in brain homogenates devoid of tau pathology. This unexpected binding was demonstrated to be because of nanomolar affinities of [3 H]AV-1451 for monoamine oxidase A and B enzymes. CONCLUSIONS: High affinity of AV-1451 for monoamine oxidase proteins may limit its utility as a tau PET tracer in PSP and Alzheimer's disease because of high levels of monoamine oxidase expression in brain regions also affected by tau deposition, especially if monoamine oxidase levels change over time or with a treatment intervention. © 2017 International Parkinson and Movement Disorder Society.


Assuntos
Encéfalo , Carbolinas/farmacocinética , Meios de Contraste/farmacocinética , Monoaminoxidase/efeitos dos fármacos , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Relação Dose-Resposta a Droga , Humanos , Tomografia por Emissão de Pósitrons , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Paralisia Supranuclear Progressiva/metabolismo , Paralisia Supranuclear Progressiva/patologia , Trítio/farmacocinética
11.
Curr Opin Neurobiol ; 27: 110-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24709372

RESUMO

Alzheimer's disease (AD) is the most prevalent cause of dementia, affecting more than 25 million people worldwide. Current models of the pathophysiological mechanisms of AD suggest that the accumulation of soluble oligomeric forms of amyloid-ß (Aß) peptides causes early loss of excitatory synapses and impairs synaptic plasticity. The signaling pathways mediating Aß oligomer-induced impairment of synaptic plasticity and loss of excitatory synapses are only beginning to be unraveled. Here, we review recent evidence supporting the critical contribution of conserved 'stress-response' kinase pathways in AD progression.


Assuntos
Doença de Alzheimer/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Progressão da Doença , Humanos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
12.
Neuron ; 78(1): 94-108, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23583109

RESUMO

Amyloid-ß 1-42 (Aß42) oligomers are synaptotoxic for excitatory cortical and hippocampal neurons and might play a role in early stages of Alzheimer's disease (AD) progression. Recent results suggested that Aß42 oligomers trigger activation of AMP-activated kinase (AMPK), and its activation is increased in the brain of patients with AD. We show that increased intracellular calcium [Ca²âº](i) induced by NMDA receptor activation or membrane depolarization activates AMPK in a CAMKK2-dependent manner. CAMKK2 or AMPK overactivation is sufficient to induce dendritic spine loss. Conversely, inhibiting their activity protects hippocampal neurons against synaptotoxic effects of Aß42 oligomers in vitro and against the loss of dendritic spines observed in the human APP(SWE,IND)-expressing transgenic mouse model in vivo. AMPK phosphorylates Tau on KxGS motif S262, and expression of Tau S262A inhibits the synaptotoxic effects of Aß42 oligomers. Our results identify a CAMKK2-AMPK-Tau pathway as a critical mediator of the synaptotoxic effects of Aß42 oligomers.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Neurônios/fisiologia , Fragmentos de Peptídeos/toxicidade , Proteínas Quinases/metabolismo , Proteínas tau/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/citologia , Cálcio/metabolismo , Células Cultivadas , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/genética , Relação Dose-Resposta a Droga , Eletroporação , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/genética , Serina/metabolismo , Transfecção
13.
Development ; 139(3): 475-87, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22223678

RESUMO

During cerebral cortex development, precise control of precursor cell cycle length and cell cycle exit is required for balanced precursor pool expansion and layer-specific neurogenesis. Here, we defined the roles of cyclin-dependent kinase inhibitor (CKI) p57(KIP2), an important regulator of G1 phase, using deletion mutant mice. Mutant mice displayed macroencephaly associated with cortical hyperplasia during late embryogenesis and postnatal development. Embryonically, proliferation of radial glial cells (RGC) and intermediate precursors (IPC) was increased, expanding both populations, with greater effect on IPCs. Furthermore, cell cycle re-entry was increased during early corticogenesis, whereas cell cycle exit was augmented at middle stage. Consequently, neurogenesis was reduced early, whereas it was enhanced during later development. In agreement, the timetable of early neurogenesis, indicated by birthdating analysis, was delayed. Cell cycle dynamics analyses in mutants indicated that p57(KIP2) regulates cell cycle length in both RGCs and IPCs. By contrast, related CKI p27(KIP1) controlled IPC proliferation exclusively. Furthermore, p57(KIP2) deficiency markedly increased RGC and IPC divisions at E14.5, whereas p27(KIP1) increased IPC proliferation at E16.5. Consequently, loss of p57(KIP2) increased primarily layer 5-6 neuron production, whereas loss of p27(KIP1) increased neurons specifically in layers 2-5. In conclusion, our observations suggest that p57(KIP2) and p27(KIP1) control neuronal output for distinct cortical layers by regulating different stages of precursor proliferation, and support a model in which IPCs contribute to both lower and upper layer neuron generation.


Assuntos
Ciclo Celular , Córtex Cerebral/crescimento & desenvolvimento , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Neurogênese , Neuroglia/metabolismo , Células-Tronco/metabolismo , Animais , Encéfalo/anormalidades , Encéfalo/crescimento & desenvolvimento , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p57/genética , Feminino , Camundongos , Deleção de Sequência
14.
Dev Neurobiol ; 72(6): 821-42, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22076965

RESUMO

The members of the CIP/KIP family of cyclin-dependent kinase (CDK) inhibitory proteins (CKIs), including p57(KIP2), p27(KIP1), and p21(CIP1), block the progression of the cell cycle by binding and inhibiting cyclin/CDK complexes of the G1 phase. In addition to this well-characterized function, p57(KIP2) and p27(KIP1) have been shown to participate in an increasing number of other important cellular processes including cell fate and differentiation, cell motility and migration, and cell death/survival, both in peripheral and central nervous systems. Increasing evidence over the past few years has characterized the functions of the newest CIP/KIP member p57(KIP2) in orchestrating cell proliferation, differentiation, and migration during neurogenesis. Here, we focus our discussion on the multiple roles played by p57(KIP2) during cortical development, making comparisons to p27(KIP1) as well as the INK4 family of CKIs.


Assuntos
Córtex Cerebral/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Animais , Ciclo Celular/fisiologia , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Córtex Cerebral/citologia , Neurônios/citologia
15.
Cereb Cortex ; 21(8): 1840-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21245411

RESUMO

Mounting evidence indicates cyclin-dependent kinase (CDK) inhibitors (CKIs) of the Cip/Kip family, including p57(Kip2) and p27(Kip1), control not only cell cycle exit but also corticogenesis. Nevertheless, distinct activities of p57(Kip2) remain poorly defined. Using in vivo and culture approaches, we show p57(Kip2) overexpression at E14.5-15.5 elicits precursor cell cycle exit, promotes transition from proliferation to neuronal differentiation, and enhances process outgrowth, while opposite effects occur in p57(Kip2)-deficient precursors. Studies at later ages indicate p57(Kip2) overexpression also induces precocious glial differentiation, suggesting stage-dependent effects. In embryonic cortex, p57(Kip2) overexpression advances cell radial migration and alters postnatal laminar positioning. While both CKIs induce differentiation, p57(Kip2) was twice as effective as p27(Kip1) in inducing neuronal differentiation and was not permissive to astrogliogenic effects of ciliary neurotrophic factor, suggesting that the CKIs differentially modulate cell fate decisions. At molecular levels, although highly conserved N-terminal regions of both CKIs elicit cycle withdrawal and differentiation, the C-terminal region of p57(Kip2) alone inhibits in vivo migration. Furthermore, p57(Kip2) effects on neurogenesis and gliogenesis require the N-terminal cyclin/CDK binding/inhibitory domains, while previous p27(Kip1) studies report cell cycle-independent functions. These observations suggest p57(Kip2) coordinates multiple stages of corticogenesis and exhibits distinct and common activities compared with related family member p27(Kip1).


Assuntos
Diferenciação Celular/fisiologia , Inibição de Migração Celular/fisiologia , Córtex Cerebral/embriologia , Inibidor de Quinase Dependente de Ciclina p57/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Neurais/enzimologia , Neurogênese/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Inibidor de Quinase Dependente de Ciclina p57/deficiência , Inibidor de Quinase Dependente de Ciclina p57/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Gravidez , Ratos , Ratos Sprague-Dawley
16.
J Neurosci ; 29(3): 775-88, 2009 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19158303

RESUMO

Although survival-promoting effects of insulin-like growth factor-1 (IGF-1) during neurogenesis are well characterized, mitogenic effects remain less well substantiated. Here, we characterize cell cycle regulators and signaling pathways underlying IGF-1 effects on embryonic cortical precursor proliferation in vitro and in vivo. In vitro, IGF-1 stimulated cell cycle progression and increased cell number without promoting cell survival. IGF-1 induced rapid increases in cyclin D1 and D3 protein levels at 4 h and cyclin E at 8 h. Moreover, p27(KIP1) and p57(KIP2) expression were reduced, suggesting downregulation of negative regulators contributes to mitogenesis. Furthermore, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway specifically underlies IGF-1 activity, because blocking this pathway, but not MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal-regulated kinase), prevented mitogenesis. To determine whether mechanisms defined in culture relate to corticogenesis in vivo, we performed transuterine intracerebroventricular injections. Whereas blockade of endogenous factor with anti-IGF-1 antibody decreased DNA synthesis, IGF-1 injection stimulated DNA synthesis and increased the number of S-phase cells in the ventricular zone. IGF-1 treatment increased phospho-Akt fourfold at 30 min, cyclins D1 and E by 6 h, and decreased p27(KIP1) and p57(KIP2) expression. Moreover, blockade of the PI3K/Akt pathway in vivo decreased DNA synthesis and cyclin E, increased p27(KIP1) and p57(KIP2) expression, and prevented IGF-1-induced cyclin E mRNA upregulation. Finally, IGF-1 injection in embryos increased postnatal day 10 brain DNA content by 28%, suggesting a role for IGF-1 in brain growth control. These results demonstrate a mitogenic role for IGF-1 that tightly controls both positive and negative cell cycle regulators, and indicate that the PI3K/Akt pathway mediates IGF-1 mitogenic signaling during corticogenesis.


Assuntos
Ciclo Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Ciclinas/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/farmacologia , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Técnicas In Vitro , Injeções Intraventriculares/métodos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Gravidez , Ratos , Transdução de Sinais/efeitos dos fármacos , Timidina/metabolismo , Fatores de Tempo
17.
Dev Neurobiol ; 69(1): 1-21, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18814313

RESUMO

In developing central nervous system, a variety of mechanisms couple cell cycle exit to differentiation during neurogenesis. The cyclin-dependent kinase (CDK) inhibitor p57Kip2 controls the transition from proliferation to differentiation in many tissues, but roles in developing brain remain uncertain. To characterize possible functions, we defined p57Kip2 protein expression in embryonic (E) day 12.5 to 20.5 rat brains using immunohistochemistry combined with markers of proliferation and differentiation. The p57Kip2 was localized primarily in cell nuclei and positive cells formed two distinct patterns including wide dispersion and laminar aggregation that were brain region-specific. From E12.5 to E16.5, p57Kip2 expression was detected mainly in ventricular zone (VZ) and/or mantle zone of hippocampus, septum, basal ganglia, thalamus, hypothalamus, midbrain, and spinal cord. After E18.5, p57Kip2 was detected in select regions undergoing differentiation. The p57Kip2 expression was also compared with regional transcription factors, including Ngn2, Nkx2.1, and Pax6. Time course studies performed in diencephalon showed that p57Kip2 immunoreactivity colocalized with BrdU at 8 hr in nuclei exhibiting the wide dispersion pattern, whereas colocalization in the laminar pattern occurred only later. Moreover, p57Kip2 frequently colocalized with neuronal marker, beta-III tubulin. Finally, we characterized relationships of p57Kip2 to CDK inhibitor p27Kip1: in proliferative regions, p57Kip2 expression preceded p27Kip1 as cells underwent differentiation, though the proteins colocalized in substantial numbers of cells, suggesting potentially related yet distinct functions of Cip/Kip family members during neurogenesis. Our observations that p57Kip2 exhibits nuclear expression as precursors exit the cell cycle and begin expressing neuronal characteristics suggests that the CDK inhibitor contributes to regulating the transition from proliferation to differentiation during brain development.


Assuntos
Encéfalo , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Células-Tronco Embrionárias/fisiologia , Neurônios/fisiologia , Animais , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/metabolismo , Bromodesoxiuridina/metabolismo , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p57/genética , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurogênese/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
18.
Histochem Cell Biol ; 127(2): 195-203, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17024454

RESUMO

As a member of the CIP/KIP family of cyclin-dependent kinase inhibitors (CKIs), p57Kip2 binds tightly to G1 cyclin/cyclin-dependent kinase complexes to block cell cycle progression. CKIs play critical roles in regulating the transition from proliferation to differentiation in many tissues, including the nervous system. Conversely, CKI dys-regulation contributes to neoplasia and cancer progression. While the combined detection of CKI immunoreactivity and S phase entry using bromodeoxyuridine (BrdU) incorporation may be particularly informative, successful immunostaining may be limited due to "masked" antigen epitopes and acid-induced signal degradation. We now report an improved double immunofluorescent method for detecting p57Kip2 and BrdU in paraformaldehyde-fixed frozen sections of embryonic rat brain. We substituted deoxyribonuclease I (DNAse I) for HCl pre-treatment to expose antigenic sites in frozen sections, and employed a biotinylated tyramide-based system to enhance p57Kip2 visualization. We identified a time- and dose-dependent relationship between DNAse I treatment and double labeling of p57Kip2 and BrdU, increasing both the numbers and intensities of immunopositive nuclei. With excess DNAse I treatment, however, there was signal degradation for both BrdU and total DNA, as reflected by DAPI staining. The use of DNAse I pre-treatment significantly increases the reliability and sensitivity of immunodetection of CKI nuclear factors, and should be useful for both developmental neurobiology studies as well as cancer diagnostic applications.


Assuntos
Química Encefálica , Encéfalo/embriologia , Bromodesoxiuridina/análise , Inibidor de Quinase Dependente de Ciclina p57/análise , Desoxirribonuclease I/metabolismo , Imuno-Histoquímica/métodos , Animais , Encéfalo/citologia , Bromodesoxiuridina/isolamento & purificação , Inibidor de Quinase Dependente de Ciclina p57/isolamento & purificação , Embrião de Mamíferos , Feminino , Gravidez , Ratos , Ratos Sprague-Dawley
19.
Biochim Biophys Acta ; 1759(5): 225-33, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16806532

RESUMO

Flavoproteins of the quiescin/sulfhydryl oxidase (QSOX) family catalyze oxidation of peptide and protein thiols to disulfides with the reduction of oxygen to hydrogen peroxide. We report here the molecular cloning of a new putative sulfhydryl oxidase cDNA, rQSOX-L (GenBank Accession no ), from adult rat brain and its expression studied by RT-PCR, Northern and Western blots in rat tissues. DNA-sequencing demonstrated the existence of two cDNAs in rat cortex, corresponding to a long transcript (rQSOX-L) and a short transcript (rQSOX-S) which differed by 851 nucleotides due to alternative splicing. The new transcript, rQSOX-L (3356 nucleotides), was specifically expressed in brain, hypophysis, heart, testis and seminal vesicle. The distribution of this variant is not homogeneous in the different tissues studied and suggests a complex gene regulation. The full-length rQSOX-L cDNA has an open reading frame of 2250-bp encoding a protein of 750 amino acids that contains a signal peptide sequence, a protein-disulfide-isomerase-type thioredoxin and ERV1-ALR domains and a long form specific C-terminal extension. The rQSOX-L protein is highly homologous to members of the sulfhydryl oxidase/Quiescin family and contains particularly two potential sites for N-glycosylation. This protein isoform was specifically detected in rat brain tissues in opposition to the low molecular form that was ubiquitous. Matrix-assisted laser desorption/ionization time of flight mass spectrometry analysis of the immunoprecipitate tryptic fragments allowed the identification of rQSOX-L protein.


Assuntos
Processamento Alternativo , Córtex Cerebral/enzimologia , Oxirredutases/genética , Sequência de Aminoácidos , Animais , Encéfalo/enzimologia , Flavina-Adenina Dinucleotídeo/metabolismo , Expressão Gênica , Genoma , Glicosilação , Imunoprecipitação , Masculino , Dados de Sequência Molecular , Oxirredutases/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transcrição Gênica
20.
Cell Tissue Res ; 323(1): 91-103, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16160860

RESUMO

Rat quiescin/sulphydryl oxidase (rQSOX) introduces disulphide bridges into peptides and proteins with the reduction of molecular oxygen to hydrogen peroxide. Its occurrence has been previously highlighted in a wide range of organs by reverse transcription-polymerase chain reaction (RT-PCR) and Northern blot analyses, methods that have provided information concerning its expression in whole organs but that do not reveal the cell types expressing this enzyme. In this report, in addition to RT-PCR and Western blot experiments, the cell-specific localization of rQSOX has been investigated in a wide range of male and female adult rat tissues by using in situ hybridization and immunohistochemistry. Labelling was detected in most organs and systems including the immune, endocrine and reproductive systems, the respiratory, digestive and urinary tracts and the skin. No labelling was observed in the heart, blood vessel endothelium, liver or smooth and skeletal muscles. rQSOX expression was mainly localized in epithelial cells specialized in secretion, strengthening the hypothesis that QSOX enzymes play an important role in the mechanism of secretion, notably in the folding of secreted proteins. The intracellular patterns of immunolabelling indicate that the protein usually follows the secretory pathway, which is in accordance with its secreted nature and its presumed involvement in the elaboration of the extracellular matrix. In seminiferous tubules, where a high level of expression was noticed, QSOX might play an important physiological role in sperm function and serve as a marker for the diagnosis of male infertility.


Assuntos
Regulação da Expressão Gênica , Oxirredutases/metabolismo , Túbulos Seminíferos/metabolismo , Animais , Feminino , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...