Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Glia ; 61(6): 869-80, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23483656

RESUMO

Oligodendrocytes make myelin for rapid impulse propagation and contribute to the long-term survival of myelinated axons. The mechanisms by which oligodendroglial dysfunction(s) contribute to slowly progressive neurodegeneration are not well understood. Here, we demonstrate in Cnp1 mutant mice that secondary axonal degeneration in the subcortical white matter is associated with an age-dependent activation of both, innate and adaptive immune responses, including an expansion of infiltrating CD8+ T cells. While the detrimental role of lymphocytes in inherited myelin diseases is known, the role of activated microglia for the hypothetical cycle of inflammation/degeneration is unclear. We used a mild standardized cryolesion of the right parietal cortex to activate microglia at the vulnerable age of mouse puberty (postnatal day (P) 28). When applied to Cnp1 mutant mice, analyzed more than 3 months later, minor brain injury had acted as a "second hit" and significantly enhanced astrogliosis, microgliosis and axon degeneration, but not T cell infiltration. Interestingly, exacerbated neuropathological changes were also reflected by specific deterioration of working memory on top of an essentially normal basic behavior. We propose a model in which oligodendroglial dysfunctions can trigger a vicious cycle of neurodegeneration and low-grade inflammation that is amplified by nonspecific activators of the innate immune system. This interaction of genetic and environmental factors may be relevant for neuropsychiatric diseases associated with secondary neuroinflammation.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/metabolismo , Axônios/metabolismo , Lesões Encefálicas/metabolismo , Degeneração Neural/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , Animais , Axônios/patologia , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Gliose/metabolismo , Gliose/patologia , Gliose/fisiopatologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Atividade Motora/fisiologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Fibras Nervosas Mielinizadas/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Teste de Desempenho do Rota-Rod , Comportamento Social , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia
2.
BMC Biol ; 6: 37, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18782446

RESUMO

BACKGROUND: Erythropoietin (EPO) improves cognition of human subjects in the clinical setting by as yet unknown mechanisms. We developed a mouse model of robust cognitive improvement by EPO to obtain the first clues of how EPO influences cognition, and how it may act on hippocampal neurons to modulate plasticity. RESULTS: We show here that a 3-week treatment of young mice with EPO enhances long-term potentiation (LTP), a cellular correlate of learning processes in the CA1 region of the hippocampus. This treatment concomitantly alters short-term synaptic plasticity and synaptic transmission, shifting the balance of excitatory and inhibitory activity. These effects are accompanied by an improvement of hippocampus dependent memory, persisting for 3 weeks after termination of EPO injections, and are independent of changes in hematocrit. Networks of EPO-treated primary hippocampal neurons develop lower overall spiking activity but enhanced bursting in discrete neuronal assemblies. At the level of developing single neurons, EPO treatment reduces the typical increase in excitatory synaptic transmission without changing the number of synaptic boutons, consistent with prolonged functional silencing of synapses. CONCLUSION: We conclude that EPO improves hippocampus dependent memory by modulating plasticity, synaptic connectivity and activity of memory-related neuronal networks. These mechanisms of action of EPO have to be further exploited for treating neuropsychiatric diseases.


Assuntos
Eritropoetina/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Eletrofisiologia , Hipocampo/fisiologia , Immunoblotting , Potenciação de Longa Duração/fisiologia , Masculino , Memória/fisiologia , Camundongos , Microscopia Confocal , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/fisiologia
3.
EMBO Mol Med ; 3(6): 309-19, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21433290

RESUMO

KCNN3, encoding the small conductance calcium-activated potassium channel SK3, harbours a polymorphic CAG repeat in the amino-terminal coding region with yet unproven function. Hypothesizing that KCNN3 genotypes do not influence susceptibility to schizophrenia but modify its phenotype, we explored their contribution to specific schizophrenic symptoms. Using the Göttingen Research Association for Schizophrenia (GRAS) data collection of schizophrenic patients (n = 1074), we performed a phenotype-based genetic association study (PGAS) of KCNN3. We show that long CAG repeats in the schizophrenic sample are specifically associated with better performance in higher cognitive tasks, comprising the capacity to discriminate, select and execute (p < 0.0001). Long repeats reduce SK3 channel function, as we demonstrate by patch-clamping of transfected HEK293 cells. In contrast, modelling the opposite in mice, i.e. KCNN3 overexpression/channel hyperfunction, leads to selective deficits in higher brain functions comparable to those influenced by SK3 conductance in humans. To conclude, KCNN3 genotypes modify cognitive performance, shown here in a large sample of schizophrenic patients. Reduction of SK3 function may constitute a pharmacological target to improve cognition in schizophrenia and other conditions with cognitive impairment.


Assuntos
Esquizofrenia/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Repetições de Trinucleotídeos/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Animais , Sequência de Bases , Estudos de Casos e Controles , Linhagem Celular , Cognição/fisiologia , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Técnicas de Patch-Clamp , Fenótipo , Polimorfismo Genético , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo
4.
Behav Brain Res ; 208(1): 80-4, 2010 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19900484

RESUMO

We have previously shown that high-dose erythropoietin (EPO) treatment improves hippocampal plasticity and cognitive performance in rodents and in patients suffering from neuropsychiatric diseases. It was therefore of interest to explore whether upregulation of endogenous EPO in brain by hypoxia inducible factor (HIF) stabilization would increase hippocampal memory similar to exogenous EPO. HIFs are transcription factors involved in the cellular response to low oxygen, including upregulation of transcripts like vascular endothelial growth factor (VEGF) and EPO. Under normal oxygen, prolylhydroxylases decrease HIF-alpha stability. This is banned by prolylhydroxylase inhibitors, which prevent oxygen dependent degradation and thus prolong HIF-alpha half life. In an experimental set-up identical to the one yielding strong cognitive effects with EPO, healthy male 28-day-old mice received FG-4497, a HIF prolylhydroxylase inhibitor, or placebo intraperitoneally every other day for 3 weeks. Behavioral testing and hematocrit determinations were conducted in independent cohorts at 1, 3, or 4 weeks after treatment completion. Increased EPO and VEGF mRNA expression in hippocampus or primary hippocampal neurons 6h after the application of FG-4497 confirmed its ability to stabilize HIF and upregulate HIF dependent transcription in brain. At 3 and 4 weeks after the last injection, respectively, FG-4497 treated mice compared to placebo mice had improved hippocampal memory in fear conditioning without change in hematocrit. In contrast, no improvement in memory was detected at 1 week, when the hematocrit was increased, indicating that cognitive improvement and hematocrit are not directly related. FG-4497 application for 3 weeks leads to delayed but lasting enhancement of hippocampal memory, making HIF stabilization an attractive target for pharmacological manipulation of cognition.


Assuntos
Hipocampo/fisiologia , Fator 1 Induzível por Hipóxia/metabolismo , Memória/efeitos dos fármacos , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal , Células Cultivadas , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Inibidores Enzimáticos/farmacologia , Eritropoetina/genética , Eritropoetina/metabolismo , Medo , Hematócrito/métodos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Behav Brain Res ; 204(1): 246-9, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19482043

RESUMO

Chronic psychosocial stress has been suggested as "second hit" in the etiology of neuropsychiatric disease, but experimental evidence is scarce. We employed repetitive social defeat stress in juvenile mice, housed individually or in groups, and measured sensorimotor gating by pre-pulse inhibition (PPI), a marker of neuronal network function. Using the resident-intruder paradigm, 28-day old C57BL/6NCrl mice were subjected daily for 3 weeks to social defeat. PPI and basic behaviour were analyzed 10 weeks later. Whereas stress increased the level of anxiety in all animals, persistent PPI deficits were found only in individually housed mice. Thus, social support in situations of severe psychosocial stress may prevent lasting impairment in basic information processing.


Assuntos
Comportamento Impulsivo , Isolamento Social , Estresse Psicológico , Análise de Variância , Animais , Ansiedade/psicologia , Abrigo para Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Testes Neuropsicológicos , Comportamento Social , Apoio Social , Fatores de Tempo
6.
Behav Pharmacol ; 17(4): 295-302, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16914947

RESUMO

Previous studies suggested that adenosine A1 and A2A receptor agonists counteract behavioral effects of N-methyl-D-aspartate (NMDA) receptor antagonists while adenosine receptor antagonists may produce opposite effects enhancing the actions of NMDA receptor antagonists. To further evaluate the effects of combined administration of adenosine receptor antagonist caffeine and various NMDA and non-NMDA glutamate receptor antagonists on brain stimulation reward (discrete-trial threshold current intensity titration procedure), rats with electrodes implanted into the ventral tegmental area were tested after pretreatment with NMDA receptor channel blocker MK-801 (0.01-0.3 mg/kg), competitive antagonist D-CPPene (0.3-5.6 mg/kg), glycine site antagonist L-701,324 (1.25-5 mg/kg), alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor antagonist GYKI-53655 (1-10 mg/kg), metabotropic glutamate receptor 5 (mGluR5) antagonist MPEP (1-10 mg/kg) alone and in combination with caffeine (1-30 mg/kg). MK-801 (0.056 and 0.1 mg/kg) was the only tested glutamate antagonist that lowered self-stimulation thresholds, while D-CPPene (5.6 mg/kg) and MPEP (5.6 and 10 mg/kg) had the opposite effects. Threshold-increasing effects of D-CPPene, but not of MPEP, however, were associated with marked impairment of operant performance, reflected by longer latencies to respond and higher rates of responding during the inter-trial intervals. Operant performance was also disrupted by the highest dose of MK-801 (0.3 mg/kg). For subsequent experiments, caffeine (1-30 mg/kg) was combined with the highest doses of NMDA receptor antagonists that did not lower the brain stimulation reward thresholds and did not impair operant performance. Caffeine had no appreciable effects on self-stimulation behavior when given alone. A low dose of caffeine (3 mg/kg) significantly lowered self-stimulation thresholds only when given together with MK-801 (0.03 mg/kg) or D-CPPene (3 mg/kg). Combined with the same antagonist drugs, higher doses of caffeine (10 and 30 mg/kg) facilitated time-out responding. These results indicate that, within a limited dose range, caffeine in combination with an NMDA receptor channel blocker and a competitive antagonist significantly lowers brain stimulation reward thresholds in rats.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , N-Metilaspartato/farmacologia , Receptores de AMPA/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Recompensa , Animais , Benzodiazepinas/farmacologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Estimulação Elétrica , Masculino , Piperazinas/farmacologia , Piridinas/farmacologia , Quinolonas/farmacologia , Ratos , Ratos Wistar , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/fisiologia , Receptor de Glutamato Metabotrópico 5 , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Autoestimulação , Área Tegmentar Ventral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA