Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Neuroinflammation ; 17(1): 159, 2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32429943

RESUMO

BACKGROUND: Cosmic radiation exposures have been found to elicit cognitive impairments involving a wide-range of underlying neuropathology including elevated oxidative stress, neural stem cell loss, and compromised neuronal architecture. Cognitive impairments have also been associated with sustained microglia activation following low dose exposure to helium ions. Space-relevant charged particles elicit neuroinflammation that persists long-term post-irradiation. Here, we investigated the potential neurocognitive benefits of microglia depletion following low dose whole body exposure to helium ions. METHODS: Adult mice were administered a dietary inhibitor (PLX5622) of colony stimulating factor-1 receptor (CSF1R) to deplete microglia 2 weeks after whole body helium irradiation (4He, 30 cGy, 400 MeV/n). Cohorts of mice maintained on a normal and PLX5622 diet were tested for cognitive function using seven independent behavioral tasks, microglial activation, hippocampal neuronal morphology, spine density, and electrophysiology properties 4-6 weeks later. RESULTS: PLX5622 treatment caused a rapid and near complete elimination of microglia in the brain within 3 days of treatment. Irradiated animals on normal diet exhibited a range of behavioral deficits involving the medial pre-frontal cortex and hippocampus and increased microglial activation. Animals on PLX5622 diet exhibited no radiation-induced cognitive deficits, and expression of resting and activated microglia were almost completely abolished, without any effects on the oligodendrocyte progenitors, throughout the brain. While PLX5622 treatment was found to attenuate radiation-induced increases in post-synaptic density protein 95 (PSD-95) puncta and to preserve mushroom type spine densities, other morphologic features of neurons and electrophysiologic measures of intrinsic excitability were relatively unaffected. CONCLUSIONS: Our data suggest that microglia play a critical role in cosmic radiation-induced cognitive deficits in mice and, that approaches targeting microglial function are poised to provide considerable benefit to the brain exposed to charged particles.


Assuntos
Encéfalo/efeitos da radiação , Hélio/toxicidade , Microglia , Lesões Experimentais por Radiação/patologia , Animais , Disfunção Cognitiva/etiologia , Radiação Cósmica/efeitos adversos , Masculino , Camundongos
2.
Cereb Cortex ; 28(12): 4454-4471, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30307491

RESUMO

Neural stem cells (NSCs) in the adult mouse hippocampus occur in a specific neurogenic niche, where a multitude of extracellular signaling molecules converges to regulate NSC proliferation as well as fate and functional integration. However, the underlying mechanisms how NSCs react to extrinsic signals and convert them to intracellular responses still remains elusive. NSCs contain a functional endocannabinoid system, including the cannabinoid type-1 receptor (CB1). To decipher whether CB1 regulates adult neurogenesis directly or indirectly in vivo, we performed NSC-specific conditional inactivation of CB1 by using triple-transgenic mice. Here, we show that lack of CB1 in NSCs is sufficient to decrease proliferation of the stem cell pool, which consequently leads to a reduction in the number of newborn neurons. Furthermore, neuronal differentiation was compromised at the level of dendritic maturation pointing towards a postsynaptic role of CB1 in vivo. Deteriorated neurogenesis in NSC-specific CB1 knock-outs additionally resulted in reduced long-term potentiation in the hippocampal formation. The observed cellular and physiological alterations led to decreased short-term spatial memory and increased depression-like behavior. These results demonstrate that CB1 expressed in NSCs and their progeny controls neurogenesis in adult mice to regulate the NSC stem cell pool, dendritic morphology, activity-dependent plasticity, and behavior.


Assuntos
Hipocampo/fisiologia , Potenciação de Longa Duração , Células-Tronco Neurais/fisiologia , Neurogênese , Receptor CB1 de Canabinoide/fisiologia , Animais , Comportamento Animal , Hipocampo/citologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Receptor CB1 de Canabinoide/genética , Memória Espacial/fisiologia
3.
J Neurosci ; 37(43): 10278-10289, 2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-28924012

RESUMO

Creutzfeldt-Jakob disease (CJD) is a neurodegenerative disorder caused by prion protein (PrP) misfolding, clinically recognized by cognitive and motor deficits, electroencephalographic abnormalities, and seizures. Its neurophysiological bases are not known. To assess the potential involvement of NMDA receptor (NMDAR) dysfunction, we analyzed NMDA-dependent synaptic plasticity in hippocampal slices from Tg(CJD) mice, which model a genetic form of CJD. Because PrP depletion may result in functional upregulation of NMDARs, we also analyzed PrP knock-out (KO) mice. Long-term potentiation (LTP) at the Schaffer collateral-commissural synapses in the CA1 area of ∼100-d-old Tg(CJD) mice was comparable to that of wild-type (WT) controls, but there was an inversion of metaplasticity, with increased GluN2B phosphorylation, which is indicative of enhanced NMDAR activation. Similar but less marked changes were seen in PrP KO mice. At ∼300 d of age, the magnitude of LTP increased in Tg(CJD) mice but decreased in PrP KO mice, indicating divergent changes in hippocampal synaptic responsiveness. Tg(CJD) but not PrP KO mice were intrinsically more susceptible than WT controls to focal hippocampal seizures induced by kainic acid. IL-1ß-positive astrocytes increased in the Tg(CJD) hippocampus, and blocking IL-1 receptor signaling restored normal synaptic responses and reduced seizure susceptibility. These results indicate that alterations in NMDA-dependent glutamatergic transmission in Tg(CJD) mice do not depend solely on PrP functional loss. Moreover, astrocytic IL-1ß plays a role in the enhanced synaptic responsiveness and seizure susceptibility, suggesting that targeting IL-1ß signaling may offer a novel symptomatic treatment for CJD.SIGNIFICANCE STATEMENT Dementia and myoclonic jerks develop in individuals with Creutzfeldt-Jakob disease (CJD), an incurable brain disorder caused by alterations in prion protein structure. These individuals are prone to seizures and have high brain levels of the inflammatory cytokine IL-1ß. Here we show that blocking IL-1ß receptors with anakinra, the human recombinant form of the endogenous IL-1 receptor antagonist used to treat rheumatoid arthritis, normalizes hippocampal neurotransmission and reduces seizure susceptibility in a CJD mouse model. These results link neuroinflammation to defective neurotransmission and the enhanced susceptibility to seizures in CJD and raise the possibility that targeting IL-1ß with clinically available drugs may be beneficial for symptomatic treatment of the disease.


Assuntos
Síndrome de Creutzfeldt-Jakob/tratamento farmacológico , Modelos Animais de Doenças , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Interleucina-1beta/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Convulsões/tratamento farmacológico , Animais , Síndrome de Creutzfeldt-Jakob/metabolismo , Suscetibilidade a Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , N-Metilaspartato/antagonistas & inibidores , N-Metilaspartato/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Distribuição Aleatória , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
5.
Science ; 383(6682): 491-493, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38300998

RESUMO

Highlights from the Science family of journals.

6.
7.
8.
Science ; 384(6696): 632-634, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38723087

RESUMO

Highlights from the Science family of journals.

9.
Science ; 384(6698): 871-873, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38781360

RESUMO

Highlights from the Science family of journals.

10.
Science ; 384(6699): 972-974, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38815036

RESUMO

Highlights from the Science family of journals.

11.
Science ; 384(6700): 1078-1080, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38843347

RESUMO

Highlights from the Science family of journals.

12.
Science ; 383(6688): 1190-1191, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38484047

RESUMO

Highlights from the Science family of journals.

13.
17.
Neurobiol Dis ; 58: 102-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23523633

RESUMO

Toll-like receptor 4 (TLR4) activation in neuron and astrocytes by High Mobility Group Box 1 (HMGB1) protein is a key mechanism of seizure generation. HMGB1 also activates the Receptor for Advanced Glycation Endproducts (RAGE), but it was unknown whether RAGE activation contributes to seizures or to HMGB1 proictogenic effects. We found that acute EEG seizures induced by 7ng intrahippocampal kainic acid (KA) were significantly reduced in Rage-/- mice relative to wild type (Wt) mice. The proictogenic effect of HMGB1 was decreased in Rage-/- mice, but less so, than in Tlr4-/- mice. In a mouse mesial temporal lobe epilepsy (mTLE) model, status epilepticus induced by 200ng intrahippocampal KA and the onset of the spontaneous epileptic activity were similar in Rage-/-, Tlr4-/- and Wt mice. However, the number of hippocampal paroxysmal episodes and their duration were both decreased in epileptic Rage-/- and Tlr4-/- mice vs Wt mice. All strains of epileptic mice displayed similar cognitive deficits in the novel object recognition test vs the corresponding control mice. CA1 neuronal cell loss was increased in epileptic Rage-/- vs epileptic Wt mice, while granule cell dispersion and doublecortin (DCX)-positive neurons were similarly affected. Notably, DCX neurons were preserved in epileptic Tlr4-/- mice. We did not find compensatory changes in HMGB1-related inflammatory signaling nor in glutamate receptor subunits in Rage-/- and Tlr4-/- naïve mice, except for ~20% NR2B subunit reduction in Rage-/- mice. RAGE was induced in neurons, astrocytes and microvessels in human and experimental mTLE hippocampi. We conclude that RAGE contributes to hyperexcitability underlying acute and chronic seizures, as well as to the proictogenic effects of HMGB1. RAGE and TLR4 play different roles in the neuropathologic sequelae developing after status epilepticus. These findings reveal new molecular mechanisms underlying seizures, cell loss and neurogenesis which involve inflammatory pathways upregulated in human epilepsy.


Assuntos
Epilepsia do Lobo Temporal/metabolismo , Regulação da Expressão Gênica/genética , Receptores Imunológicos/metabolismo , Convulsões/metabolismo , Regulação para Cima/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Estimulação Elétrica/efeitos adversos , Eletroencefalografia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/etiologia , Epilepsia do Lobo Temporal/patologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína HMGB1/administração & dosagem , Proteína HMGB1/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/deficiência , Convulsões/induzido quimicamente , Convulsões/etiologia , Receptor 4 Toll-Like/deficiência , Regulação para Cima/genética
18.
Science ; 379(6633): 656-657, 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36795822

RESUMO

Highlights from the Science family of journals.

19.
Science ; 381(6662): 1061-1063, 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37676937

RESUMO

Highlights from the Science family of journals.

20.
Science ; 381(6663): 1166-1167, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37708285

RESUMO

Highlights from the Science family of journals.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA