Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurodegener ; 16(1): 54, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34380535

RESUMO

BACKGROUND: Proteopathic brain lesions are a hallmark of many age-related neurodegenerative diseases including synucleinopathies and develop at least a decade before the onset of clinical symptoms. Thus, understanding of the initiation and propagation of such lesions is key for developing therapeutics to delay or halt disease progression. METHODS: Alpha-synuclein (αS) inclusions were induced in long-term murine and human slice cultures by seeded aggregation. An αS seed-recognizing human antibody was tested for blocking seeding and/or spreading of the αS lesions. Release of neurofilament light chain (NfL) into the culture medium was assessed. RESULTS: To study initial stages of α-synucleinopathies, we induced αS inclusions in murine hippocampal slice cultures by seeded aggregation. Induction of αS inclusions in neurons was apparent as early as 1week post-seeding, followed by the occurrence of microglial inclusions in vicinity of the neuronal lesions at 2-3 weeks. The amount of αS inclusions was dependent on the type of αS seed and on the culture's genetic background (wildtype vs A53T-αS genotype). Formation of αS inclusions could be monitored by neurofilament light chain protein release into the culture medium, a fluid biomarker of neurodegeneration commonly used in clinical settings. Local microinjection of αS seeds resulted in spreading of αS inclusions to neuronally connected hippocampal subregions, and seeding and spreading could be inhibited by an αS seed-recognizing human antibody. We then applied parameters of the murine cultures to surgical resection-derived adult human long-term neocortical slice cultures from 22 to 61-year-old donors. Similarly, in these human slice cultures, proof-of-principle induction of αS lesions was achieved at 1week post-seeding in combination with viral A53T-αS expressions. CONCLUSION: The successful translation of these brain cultures from mouse to human with the first reported induction of human αS lesions in a true adult human brain environment underlines the potential of this model to study proteopathic lesions in intact mouse and now even aged human brain environments.


Assuntos
Microglia/patologia , Proteínas de Neurofilamentos/metabolismo , Neurônios/patologia , Técnicas de Cultura de Órgãos/métodos , Sinucleinopatias , Animais , Humanos , Corpos de Inclusão/patologia , Camundongos , Microglia/metabolismo , Neurônios/metabolismo , alfa-Sinucleína/toxicidade
2.
Cells ; 8(9)2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31510042

RESUMO

Bone marrow Mesenchymal Stem Cells (BM-MSCs), due to their strong protective and anti-inflammatory abilities, have been widely investigated in the context of several diseases for their possible therapeutic role, based on the release of a highly proactive secretome composed of soluble factors and Extracellular Vesicles (EVs). BM-MSC-EVs, in particular, convey many of the beneficial features of parental cells, including direct and indirect ß-amyloid degrading-activities, immunoregulatory and neurotrophic abilities. Therefore, EVs represent an extremely attractive tool for therapeutic purposes in neurodegenerative diseases, including Alzheimer's disease (AD). We examined the therapeutic potential of BM-MSC-EVs injected intracerebrally into the neocortex of APPswe/PS1dE9 AD mice at 3 and 5 months of age, a time window in which the cognitive behavioral phenotype is not yet detectable or has just started to appear. We demonstrate that BM-MSC-EVs are effective at reducing the Aß plaque burden and the amount of dystrophic neurites in both the cortex and hippocampus. The presence of Neprilysin on BM-MSC-EVs, opens the possibility of a direct ß-amyloid degrading action. Our results indicate a potential role for BM-MSC-EVs already in the early stages of AD, suggesting the possibility of intervening before overt clinical manifestations.


Assuntos
Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/metabolismo , Placa Amiloide/terapia , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/metabolismo
3.
Proc Natl Acad Sci U S A ; 114(41): E8788-E8797, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-28904096

RESUMO

Dysfunction of microglia is known to play an important role in Alzheimer's disease (AD). Here, we investigated the role of RIPK1 in microglia mediating the pathogenesis of AD. RIPK1 is highly expressed by microglial cells in human AD brains. Using the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model, we found that inhibition of RIPK1, using both pharmacological and genetic means, reduced amyloid burden, the levels of inflammatory cytokines, and memory deficits. Furthermore, inhibition of RIPK1 promoted microglial degradation of Aß in vitro. We characterized the transcriptional profiles of adult microglia from APP/PS1 mice and identified a role for RIPK1 in regulating the microglial expression of CH25H and Cst7, a marker for disease-associated microglia (DAM), which encodes an endosomal/lysosomal cathepsin inhibitor named Cystatin F. We present evidence that RIPK1-mediated induction of Cst7 leads to an impairment in the lysosomal pathway. These data suggest that RIPK1 may mediate a critical checkpoint in the transition to the DAM state. Together, our study highlights a non-cell death mechanism by which the activation of RIPK1 mediates the induction of a DAM phenotype, including an inflammatory response and a reduction in phagocytic activity, and connects RIPK1-mediated transcription in microglia to the etiology of AD. Our results support that RIPK1 is an important therapeutic target for the treatment of AD.


Assuntos
Doença de Alzheimer/patologia , Biomarcadores/metabolismo , Microglia/patologia , Presenilina-1/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Fenótipo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética
4.
Sci Rep ; 7: 41734, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28134307

RESUMO

Fingolimod, also known as FTY720, is an analogue of the sphingolipid sphingosine, which has been proved to be neuroprotective in rodent models of Alzheimer's disease (AD). Several cellular and molecular targets underlying the neuroprotective effects of FTY720 have been recently identified. However, whether the drug directly protects neurons from toxicity of amyloid-beta (Aß) still remains poorly defined. Using a combination of biochemical assays, live imaging and electrophysiology we demonstrate that FTY720 induces a rapid increase in GLUN2A-containing neuroprotective NMDARs on the surface of dendritic spines in cultured hippocampal neurons. In addition, the drug mobilizes extrasynaptic GLUN2B-containing NMDARs, which are coupled to cell death, to the synapses. Altered ratio of synaptic/extrasynaptic NMDARs decreases calcium responsiveness of neurons to neurotoxic soluble Aß 1-42 and renders neurons resistant to early alteration of calcium homeostasis. The fast defensive response of FTY720 occurs through a Sphingosine-1-phosphate receptor (S1P-R) -dependent mechanism, as it is lost in the presence of S1P-R1 and S1P-R3 antagonists. We propose that rapid synaptic relocation of NMDARs might have direct impact on amelioration of cognitive performance in transgenic APPswe/PS1dE9 AD mice upon sub-chronic treatment with FTY720.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Células Piramidais/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Amiloide/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Cloridrato de Fingolimode/farmacologia , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Agregados Proteicos , Agregação Patológica de Proteínas , Ligação Proteica , Células Piramidais/efeitos dos fármacos
5.
Acta Neuropathol Commun ; 4(1): 110, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27724899

RESUMO

Substantial data indicate that amyloid-ß (Aß), the major component of senile plaques, plays a central role in Alzheimer's Disease and indeed the assembly of naturally occurring amyloid peptides into cytotoxic aggregates is linked to the disease pathogenesis. Although Aß42 is a highly aggregating form of Aß, the co-occurrence of shorter Aß peptides might affect the aggregation potential of the Aß pool. In this study we aimed to assess whether the structural behavior of human Aß42 peptide inside the brain is influenced by the concomitant presence of N-terminal fragments produced by the proteolytic activity of glial cells. We show that the occurrence of the human C-terminal truncated 1-24 Aß fragment impairs Aß42 clearance through blood brain barrier and promotes the formation of Aß42 aggregates even in the healthy brain. By showing that Aß1-24 has seeding properties for aggregate formation in intracranially injected wild type mice, our study provide the proof-of-concept that peptides produced upon Aß42 cleavage by activated glial cells may cause phenotypic defects even in the absence of genetic mutations associated with Alzheimer's Disease, possibly contributing to the development of the sporadic form of the pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Atividade Motora/fisiologia , Fragmentos de Peptídeos/administração & dosagem , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Dobramento de Proteína , Multimerização Proteica , Comportamento Social
6.
Oncotarget ; 7(5): 5143-56, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26784253

RESUMO

Astrocytes play a crucial role in neuroinflammation as part of the glia limitans, which regulates infiltration of the brain parenchyma by leukocytes. The signaling pathways and molecular events, which result from the interaction of activated T cells with astrocytes are poorly defined. Here we show that astrocytes promote the expression and enzymatic activity of CD39 and CD73 ectonucleotidases in recently activated CD4 cells by a contact dependent mechanism that is independent of T cell receptor interaction with class II major histocompatibility complex (MHC). Transforming growth factor-ß (TGF-ß) is robustly upregulated and sufficient to promote ectonucleotidases expression. T cell adhesion to astrocyte results in differentiation to an immunosuppressive phenotype defined by expression of the transcription factor Rorγt, which characterizes the CD4 T helper 17 subset. CD39 activity in T cells in turn inhibits spontaneous calcium oscillations in astrocytes that correlated with enhanced and reduced transcription of CCL2 chemokine and Sonic hedgehog (Shh), respectively. We hypothesize this TCR-independent interaction promote an immunosuppressive program in T cells to control possible brain injury by deregulated T cell activation during neuroinflammation. On the other hand, the increased secretion of CCL2 with concomitant reduction of Shh might promote leukocytes extravasation into the brain parenchyma.


Assuntos
5'-Nucleotidase/metabolismo , Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/imunologia , Astrócitos , Diferenciação Celular , Humanos , Transdução de Sinais , Regulação para Cima
7.
Biochemistry ; 51(42): 8338-52, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23025847

RESUMO

Oligomeric forms of ß-amyloid (Aß) have potent neurotoxic activity and are the primary cause of neuronal injury and cell death in Alzheimer's disease (AD). Compounds that perturb oligomer formation or structure may therefore be therapeutic for AD. We previously reported that d-[(chGly)-(Tyr)-(chGly)-(chGly)-(mLeu)]-NH(2) (SEN304) is able to inhibit Aß aggregation and toxicity, shown primarily by thioflavin T fluorescence and MTT (Kokkoni, N. et al. (2006) N-Methylated peptide inhibitors of ß-amyloid aggregation and toxicity. Optimisation of inhibitor structure. Biochemistry 45, 9906-9918). Here we extensively characterize how SEN304 affects Aß(1-42) aggregation and toxicity, using biophysical assays (thioflavin T, circular dichroism, SDS-PAGE, size exclusion chromatography, surface plasmon resonance, traveling wave ion mobility mass spectrometry, electron microscopy, ELISA), toxicity assays in cell culture (MTT and lactate dehydrogenase in human SH-SHY5Y cells, mouse neuronal cell death and synaptophysin) and long-term potentiation in a rat hippocampal brain slice. These data, with dose response curves, show that SEN304 is a powerful inhibitor of Aß(1-42) toxicity, particularly effective at preventing Aß inhibition of long-term potentiation. It can bind directly to Aß(1-42), delay ß-sheet formation and promote aggregation of toxic oligomers into a nontoxic form, with a different morphology that cannot bind thioflavin T. SEN304 appears to work by inducing aggregation, and hence removal, of Aß oligomers. It is therefore a promising lead compound for Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Multimerização Proteica/efeitos dos fármacos , Doença de Alzheimer , Animais , Benzotiazóis , Sobrevivência Celular , Dicroísmo Circular , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Estrutura Quaternária de Proteína , Ratos , Ressonância de Plasmônio de Superfície , Tiazóis , Células Tumorais Cultivadas
8.
J Neurosci ; 31(47): 16969-76, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22114267

RESUMO

Phosphorylation plays a central role in the dynamic regulation of the processing of the amyloid precursor protein (APP) and the production of amyloid-ß (Aß), one of the clinically most important factors that determine the onset of Alzheimer's disease (AD). This has led to the hypothesis that aberrant Aß production associated with AD results from regulatory defects in signal transduction. However, conflicting findings have raised a debate over the identity of the signaling pathway that controls APP metabolism. Here, we demonstrate that activation of the c-Jun N-terminal protein kinase (JNK) is essential for mediating the apoptotic response of neurons to Aß. Furthermore, we discovered that the functional loss of JNK signaling in neurons significantly decreased the number of amyloid plaques present in the brain of mice carrying familial AD-linked mutant genes. This correlated with a reduction in Aß production. Biochemical analyses indicate that the phosphorylation of APP at threonine 668 by JNK is required for γ-mediated cleavage of the C-terminal fragment of APP produced by ß-secretase. Overall, this study provides genetic evidence that JNK signaling is required for the formation of amyloid plaques in vivo. Therefore, inhibition of increased JNK activity associated with aging or with a pathological condition constitutes a potential strategy for the treatment of AD.


Assuntos
Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/deficiência , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Placa Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/genética , Células Cultivadas , Ativação Enzimática/genética , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/enzimologia , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/enzimologia , Placa Amiloide/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...