Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol Commun ; 10(1): 96, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35787306

RESUMO

Cerebrospinal fluid (CSF) biomarkers play an important role in diagnosing Alzheimer's disease (AD) which is characterized by amyloid-ß (Aß) amyloidosis. Here, we used two App knock-in mouse models, AppNL-F/NL-F and AppNL-G-F/NL-G-F, exhibiting AD-like Aß pathology to analyze how the brain pathologies translate to CSF proteomes by label-free mass spectrometry (MS). This identified several extracellular matrix (ECM) proteins as significantly altered in App knock-in mice. Next, we compared mouse CSF proteomes with previously reported human CSF MS results acquired from patients across the AD spectrum. Intriguingly, the ECM protein decorin was similarly and significantly increased in both AppNL-F/NL-F and AppNL-G-F/NL-G-F mice, strikingly already at three months of age in the AppNL-F/NL-F mice and preclinical AD subjects having abnormal CSF-Aß42 but normal cognition. Notably, in this group of subjects, CSF-decorin levels positively correlated with CSF-Aß42 levels indicating that the change in CSF-decorin is associated with early Aß amyloidosis. Importantly, receiver operating characteristic analysis revealed that CSF-decorin can predict a specific AD subtype having innate immune activation and potential choroid plexus dysfunction in the brain. Consistently, in AppNL-F/NL-F mice, increased CSF-decorin correlated with both Aß plaque load and with decorin levels in choroid plexus. In addition, a low concentration of human Aß42 induces decorin secretion from mouse primary neurons. Interestingly, we finally identify decorin to activate neuronal autophagy through enhancing lysosomal function. Altogether, the increased CSF-decorin levels occurring at an early stage of Aß amyloidosis in the brain may reflect pathological changes in choroid plexus, present in a subtype of AD subjects.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Amiloidose/patologia , Animais , Encéfalo/patologia , Decorina/líquido cefalorraquidiano , Decorina/metabolismo , Humanos , Camundongos , Placa Amiloide/patologia , Proteoma/metabolismo
2.
Sci Adv ; 8(23): eabm6155, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35675411

RESUMO

We previously developed single App knock-in mouse models of Alzheimer's disease (AD) that harbor the Swedish and Beyreuther/Iberian mutations with or without the Arctic mutation (AppNL-G-F and AppNL-F mice). We have now generated App knock-in mice devoid of the Swedish mutations (AppG-F mice) and evaluated its characteristics. Amyloid ß peptide (Aß) pathology was exhibited by AppG-F mice from 6 to 8 months of age and was accompanied by neuroinflammation. Aß-secretase inhibitor, verubecestat, attenuated Aß production in AppG-F mice, but not in AppNL-G-F mice, indicating that the AppG-F mice are more suitable for preclinical studies of ß-secretase inhibition given that most patients with AD do not carry the Swedish mutations. Comparison of isogenic App knock-in lines revealed that multiple factors, including elevated C-terminal fragment ß (CTF-ß) and humanization of Aß might influence endosomal alterations in vivo. Thus, experimental comparisons between different isogenic App, knock-in mouse lines will provide previously unidentified insights into our understanding of the etiology of AD.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Transgênicos
3.
J Biol Chem ; 297(3): 101004, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34329683

RESUMO

We previously developed single App knock-in mouse models of Alzheimer's disease (AD) harboring the Swedish and Beyreuther/Iberian mutations with or without the Arctic mutation (AppNL-G-F and AppNL-F mice, respectively). These models showed Aß pathology, neuroinflammation, and cognitive impairment in an age-dependent manner. The former model exhibits extensive pathology as early as 6 months, but is unsuitable for investigating Aß metabolism and clearance because the Arctic mutation renders Aß resistant to proteolytic degradation and prone to aggregation. In particular, it is inapplicable to preclinical immunotherapy studies due to its discrete affinity for anti-Aß antibodies. The latter model may take as long as 18 months for the pathology to become prominent, which leaves an unfulfilled need for an Alzheimer's disease animal model that is both swift to show pathology and useful for antibody therapy. We thus utilized mutant Psen1 knock-in mice into which a pathogenic mutation (P117L) had been introduced to generate a new model that exhibits early deposition of wild-type human Aß by crossbreeding the AppNL-F line with the Psen1P117L/WT line. We show that the effects of the pathogenic mutations in the App and Psen1 genes are additive or synergistic. This new third-generation mouse model showed more cored plaque pathology and neuroinflammation than AppNL-G-F mice and will help accelerate the development of disease-modifying therapies to treat preclinical AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Placa Amiloide/patologia , Animais , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Placa Amiloide/genética , Presenilina-1/genética
4.
J Alzheimers Dis ; 75(1): 321-335, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32280097

RESUMO

BACKGROUND: The 42 amino acids long amyloid-ß peptide, Aß42, may initiate a cascade of events leading to the severe neurodegeneration observed in Alzheimer's disease (AD) brain. However, the underlying molecular mechanisms remain to be established. OBJECTIVE: To find early Aß42-induced AD related mechanisms, we performed a brain proteomics time-course study on a novel App knock-in AD mouse model, AppNL-F, expressing high levels of Aß42 without AßPP overexpression artifacts. METHODS: Hippocampus and cortex were analyzed separately by using 18O-labelling mass spectrometry to reveal alterations in protein levels. Pathway analysis of proteomics data was used to identify altered biological functions. Immunohistochemistry was used to further investigate a significant key regulatory protein. RESULTS: Around 100 proteins were differently expressed in AppNL-F mice at each time point (3, 6, 9, and 18 months of age) as compared to wild type mice. Strikingly, already at 3 months of age-long before Aß plaque development and memory impairment-several pathways, including long-term potentiation and synaptic plasticity, were downregulated, and neuritogenesis was increased. Huntingtin (HTT) was identified as an upstream regulator, i.e., a key protein affecting the levels of several proteins. Increased levels of HTT in hippocampus of AppNL-F mice was supported by immunofluorescence microscopy. CONCLUSION: Notably, the proteome was significantly altered already at 3 months of age, 6 months before the development of plaques. Differentially expressed proteins varied over time, indicating that increased Aß42 levels initiate a cascade of events that eventually manifests in amyloid depositions, inflammation, and decline in memory.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Proteoma/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Córtex Cerebral/patologia , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Introdução de Genes , Hipocampo/patologia , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Transgênicos
5.
Anal Sci ; 35(4): 407-412, 2019 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-30555107

RESUMO

Racemic synephrine, which was transformed into diastereomers by derivatization with 2,3,4,6-tetra-O-acetyl-ß-D-glucopyranosil isothiocyanate, was resolved by a reversed phase HPLC with UV detection at 254 nm. The total contents of synephrine enantiomers in citrus fruit samples were exocarp > mesocarp > endocarp > sarcocarp, suggesting that synephrine content of outer side of citrus fruits was higher than that of the inner side. (R)-Synephrine was detected in exocarp of eleven fresh citrus fruits, except for lemon, lime, and grapefruit samples. (S)-Synephrine was determined in the exocarp of four citrus fruits (mikan, orange, bitter orange, and ponkan samples) and the ratio of (S)-synephrine to total synephrine was 0.5 - 0.9%. The racemization of (R)-synephrine in aqueous solution during heating at 100°C was also examined. An increase in the heating time brought about an increase in the (S)-synephrine content in a linear fashion. The racemization was found to be significantly reduced by the addition of D-fructose, D-maltose, D-glucose, D-mannose or D-galactose, but not D-sucrose or D-mannitol. It is suggested that the reducibility of sugars may result in the inhibition of racemization.


Assuntos
Citrus/química , Frutas/química , Extratos Vegetais/isolamento & purificação , Sinefrina/isolamento & purificação , Cromatografia Líquida de Alta Pressão , Estrutura Molecular , Extratos Vegetais/química , Estereoisomerismo , Sinefrina/química
6.
Nat Commun ; 9(1): 2892, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30042426

RESUMO

Base Editor (BE) and Target-AID (activation-induced cytidine deaminase) are engineered genome-editing proteins composed of Cas9 and cytidine deaminases. These base-editing tools convert C:G base pairs to T:A at target sites. Here, we inject either BE or Target-AID mRNA together with identical single-guide RNAs (sgRNAs) into mouse zygotes, and compare the base-editing efficiencies of the two distinct tools in vivo. BE consistently show higher base-editing efficiency (10.0-62.8%) compared to that of Target-AID (3.4-29.8%). However, unexpected base substitutions and insertion/deletion formations are also more frequently observed in BE-injected mice or zygotes. We are able to generate multiple mouse lines harboring point mutations in the mouse presenilin 1 (Psen1) gene by injection of BE or Target-AID. These results demonstrate that BE and Target-AID are highly useful tools to generate mice harboring pathogenic point mutations and to analyze the functional consequences of the mutations in vivo.


Assuntos
Pareamento de Bases/genética , Sistemas CRISPR-Cas , Citidina Desaminase/genética , Edição de Genes/métodos , Mutação , Presenilina-1/genética , Animais , Sequência de Bases , Citidina Desaminase/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Presenilina-1/metabolismo , Reprodutibilidade dos Testes , Homologia de Sequência do Ácido Nucleico
7.
Proteomics ; 15(19): 3349-55, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26194619

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease displaying extracellular plaques formed by the neurotoxic amyloid ß-peptide (Aß), and intracellular neurofibrillary tangles consisting of protein tau. However, how these pathologies relate to the massive neuronal death that occurs in AD brains remain elusive. Neprilysin is the major Aß-degrading enzyme and a lack thereof increases Aß levels in the brain twofold. To identify altered protein expression levels induced by increased Aß levels, we performed a proteomic analysis of the brain of the AD mouse model APPsw and compared it to that of APPsw mice lacking neprilysin. To this end we established an LC-MS/MS method to analyze brain homogenate, using an (18) O-labeled internal standard to accurately quantify the protein levels. To distinguish between alterations in protein levels caused by increased Aß levels and those induced by neprilysin deficiency independently of Aß, the brain proteome of neprilysin deficient APPsw mice was also compared to that of neprilysin deficient mice. By this approach we identified approximately 600 proteins and the levels of 300 of these were quantified. Pathway analysis showed that many of the proteins with altered expression were involved in neurological disorders, and that tau, presenilin and APP were key regulators in the identified networks. The data have been deposited to the ProteomeXchange Consortium with identifiers PXD000968 and PXD001786 (http://proteomecentral.proteomexchange.org/dataset/PXD000968 and (http://proteomecentral.proteomexchange.org/dataset/PXD001786). Interestingly, the levels of several proteins, including some not previously reported to be linked to AD, were associated with increased Aß levels.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica , Neprilisina/genética , Proteínas/genética , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiopatologia , Cromatografia Líquida , Modelos Animais de Doenças , Deleção de Genes , Camundongos , Proteínas/análise , Proteômica , Espectrometria de Massas em Tandem
8.
Am J Pathol ; 185(2): 305-13, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25433221

RESUMO

Alzheimer disease (AD) is biochemically characterized by increased levels of amyloid ß (Aß) peptide, which aggregates into extracellular Aß plaques in AD brains. Before plaque formation, Aß accumulates intracellularly in both AD brains and in the brains of AD model mice, which may contribute to disease progression. Autophagy, which is impaired in AD, clears cellular protein aggregates and participates in Aß metabolism. In addition to a degradative role of autophagy in Aß metabolism we recently showed that Aß secretion is inhibited in mice lacking autophagy-related gene 7 (Atg7) in excitatory neurons in the mouse forebrain. This inhibition of Aß secretion leads to intracellular accumulation of Aß. Here, we used fluorescence and immunoelectron microscopy to elucidate the subcellular localization of the intracellular Aß accumulation which accumulates in Aß precursor protein mice lacking Atg7. Autophagy deficiency causes accumulation of p62(+) aggregates, but these aggregates do not contain Aß. However, knockdown of Atg7 induced Aß accumulation in the Golgi and a concomitant reduction of Aß in the multivesicular bodies. This indicates that Atg7 influences the transport of Aß possibly derived from Golgi to multivesicular bodies.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Complexo de Golgi/metabolismo , Proteínas Associadas aos Microtúbulos/deficiência , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Proteína 7 Relacionada à Autofagia , Complexo de Golgi/genética , Complexo de Golgi/patologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Fragmentos de Peptídeos/genética
9.
Cell Rep ; 5(1): 61-9, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24095740

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease biochemically characterized by aberrant protein aggregation, including amyloid beta (Aß) peptide accumulation. Protein aggregates in the cell are cleared by autophagy, a mechanism impaired in AD. To investigate the role of autophagy in Aß pathology in vivo, we crossed amyloid precursor protein (APP) transgenic mice with mice lacking autophagy in excitatory forebrain neurons obtained by conditional knockout of autophagy-related protein 7. Remarkably, autophagy deficiency drastically reduced extracellular Aß plaque burden. This reduction of Aß plaque load was due to inhibition of Aß secretion, which led to aberrant intraneuronal Aß accumulation in the perinuclear region. Moreover, autophagy-deficiency-induced neurodegeneration was exacerbated by amyloidosis, which together severely impaired memory. Our results establish a function for autophagy in Aß metabolism: autophagy influences secretion of Aß to the extracellular space and thereby directly affects Aß plaque formation, a pathological hallmark of AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Autofagia/fisiologia , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia
10.
Sci Rep ; 3: 1472, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23503602

RESUMO

Accumulation of amyloid-ß peptide (Aß) in the brain is closely associated with cognitive decline in Alzheimer's disease (AD). Stereotaxic infusion of neprilysin-encoding viral vectors into the hippocampus has been shown to decrease Aß in AD-model mice, but more efficient and global delivery is necessary to treat the broadly distributed burden in AD. Here we developed an adeno-associated virus (AAV) vector capable of providing neuronal gene expression throughout the brains after peripheral administration. A single intracardiac administration of the vector carrying neprilysin gene in AD-model mice elevated neprilysin activity broadly in the brain, and reduced Aß oligomers, with concurrent alleviation of abnormal learning and memory function and improvement of amyloid burden. The exogenous neprilysin was localized mainly in endosomes, thereby effectively excluding Aß oligomers from the brain. AAV vector-mediated gene transfer may provide a therapeutic strategy for neurodegenerative diseases, where global transduction of a therapeutic gene into the brain is necessary.


Assuntos
Dependovirus/genética , Vetores Genéticos , Neprilisina/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Animais , Perfilação da Expressão Gênica , Hipocampo , Injeções Intravenosas , Camundongos , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...