Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 215(4): 1047-1058, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29483128

RESUMO

One of the hallmarks of Alzheimer's disease is the presence of extracellular diffuse and fibrillar plaques predominantly consisting of the amyloid-ß (Aß) peptide. Apolipoprotein E (ApoE) influences the deposition of amyloid pathology through affecting the clearance and aggregation of monomeric Aß in the brain. In addition to influencing Aß metabolism, increasing evidence suggests that apoE influences microglial function in neurodegenerative diseases. Here, we characterize the impact that apoE has on amyloid pathology and the innate immune response in APPPS1ΔE9 and APPPS1-21 transgenic mice. We report that Apoe deficiency reduced fibrillar plaque deposition, consistent with previous studies. However, fibrillar plaques in Apoe-deficient mice exhibited a striking reduction in plaque compaction. Hyperspectral fluorescent imaging using luminescent conjugated oligothiophenes identified distinct Aß morphotypes in Apoe-deficient mice. We also observed a significant reduction in fibrillar plaque-associated microgliosis and activated microglial gene expression in Apoe-deficient mice, along with significant increases in dystrophic neurites around fibrillar plaques. Our results suggest that apoE is critical in stimulating the innate immune response to amyloid pathology.


Assuntos
Amiloide/metabolismo , Apolipoproteínas E/metabolismo , Microglia/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/imunologia , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/imunologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Apolipoproteínas E/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Imunidade Inata/imunologia , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia , Placa Amiloide/imunologia
2.
Neuron ; 96(5): 1013-1023.e4, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29216448

RESUMO

The apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset Alzheimer disease. Previous studies suggest that reduction of apoE levels through genetic manipulation can reduce Aß pathology. However, it is not clear how reduction of apoE levels after birth would affect amyloid deposition. We utilize an antisense oligonucleotide (ASO) to reduce apoE expression in the brains of APP/PS1-21 mice homozygous for the APOE-ε4 or APOE-ε3 allele. ASO treatment starting after birth led to a significant decrease in Aß pathology when assessed at 4 months. Interestingly, ASO treatment starting at the onset of amyloid deposition led to an increase in Aß plaque size and a reduction in plaque-associated neuritic dystrophy with no change in overall plaque load. These results suggest that lowering apoE levels prior to plaque deposition can strongly affect the initiation of Aß pathology while lowering apoE after Aß seeding modulates plaque size and toxicity.


Assuntos
Peptídeos beta-Amiloides , Amiloidose/tratamento farmacológico , Apolipoproteínas E/antagonistas & inibidores , Oligonucleotídeos Antissenso/uso terapêutico , Envelhecimento/fisiologia , Alelos , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/genética , Amiloidose/patologia , Animais , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Placa Amiloide/prevenção & controle
3.
Proc Natl Acad Sci U S A ; 114(43): 11524-11529, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29073081

RESUMO

Variants in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) were recently found to increase the risk for developing Alzheimer's disease (AD). In the brain, TREM2 is predominately expressed on microglia, and its association with AD adds to increasing evidence implicating a role for the innate immune system in AD initiation and progression. Thus far, studies have found TREM2 is protective in the response to amyloid pathology while variants leading to a loss of TREM2 function impair microglial signaling and are deleterious. However, the potential role of TREM2 in the context of tau pathology has not yet been characterized. In this study, we crossed Trem2+/+ (T2+/+) and Trem2-/- (T2-/-) mice to the PS19 human tau transgenic line (PS) to investigate whether loss of TREM2 function affected tau pathology, the microglial response to tau pathology, or neurodegeneration. Strikingly, by 9 mo of age, T2-/-PS mice exhibited significantly less brain atrophy as quantified by ventricular enlargement and preserved cortical volume in the entorhinal and piriform regions compared with T2+/+PS mice. However, no TREM2-dependent differences were observed for phosphorylated tau staining or insoluble tau levels. Rather, T2-/-PS mice exhibited significantly reduced microgliosis in the hippocampus and piriform cortex compared with T2+/+PS mice. Gene expression analyses and immunostaining revealed microglial activation was significantly attenuated in T2-/-PS mice, and there were lower levels of inflammatory cytokines and astrogliosis. These unexpected findings suggest that impairing microglial TREM2 signaling reduces neuroinflammation and is protective against neurodegeneration in the setting of pure tauopathy.


Assuntos
Inflamação/genética , Glicoproteínas de Membrana/metabolismo , Doenças Neurodegenerativas/genética , Receptores Imunológicos/metabolismo , Tauopatias , Animais , Regulação da Expressão Gênica/fisiologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores Imunológicos/genética
4.
Mol Neurodegener ; 10: 55, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26502977

RESUMO

BACKGROUND: Intracellular accumulation of tau as neurofibrillary tangles (NFTs) is the hallmark of Alzheimer's disease (AD) as well as in other tauopathies. Tau is present not only in the cytoplasm but also in the extracellular space such as cerebrospinal fluid (CSF) and brain interstitial fluid (ISF). Although clearance is one critical parameter leading to such intracellular/extracellular tau accumulation, in vivo turnover of tau has not been well characterized. The current study has attempted to precisely determine in vivo turnover rates of tau utilizing tet-off regulatable mice. In particular, we assessed intracellular tau and extracellular tau, soluble tau, insoluble tau and phosphorylated tau at certain sites utilizing a combination of in vivo microdialysis, biochemical analysis and specific ELISAs recognizing each species. To examine the effect of a tauopathy-associated mutation on tau clearance, half-lives of various tau species were compared between the mice with a FTDP-17 mutation that induces ß-sheet formation, ΔK280 mutation (pro-aggregant mice) and control mice with additional ß-sheet breaking mutations (anti-aggregant mice). RESULTS: Here we report that tau is metabolized at much slower turnover rates in vivo than in cell culture. We found that insoluble tau in pro-aggregant mice had a significantly slower half-life (t1/2 = ~34.2 days) than soluble tau (t1/2 = ~9.7 days). In contrast, soluble tau phosphorylated in the proline rich region was cleared faster than total soluble tau. When comparing pro-aggregant mice to anti-agregant mice, turnover rates of soluble tau species were not significantly different. CONCLUSIONS: The current study provides a comprehensive description of in vivo turnover of various tau species present in mice that express human tau. The turnover rate of soluble tau was not significantly altered between pro-aggregant mice and anti-aggregant mice. This suggests that altered conformation by ΔK280 does not have a major impact on clearance pathways for soluble tau. In contrast, different tau species displayed different half-lives. Turnover was significantly delayed for insoluble tau whereas it was accelerated for soluble tau phosphorylated in the proline rich region. These differences in susceptibilities to clearance suggest that aggregation and phosphorylation influences tau clearance which may be important in tau pathogenesis.


Assuntos
Encéfalo/metabolismo , Demência Frontotemporal/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Tauopatias/genética
5.
J Exp Med ; 211(13): 2487-96, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25422493

RESUMO

Age-related aggregation of amyloid-ß (Aß) is an upstream pathological event in Alzheimer's disease (AD) pathogenesis, and it disrupts the sleep-wake cycle. The amount of sleep declines with aging and to a greater extent in AD. Poor sleep quality and insufficient amounts of sleep have been noted in humans with preclinical evidence of AD. However, how the amount and quality of sleep affects Aß aggregation is not yet well understood. Orexins (hypocretins) initiate and maintain wakefulness, and loss of orexin-producing neurons causes narcolepsy. We tried to determine whether orexin release or secondary changes in sleep via orexin modulation affect Aß pathology. Amyloid precursor protein (APP)/Presenilin 1 (PS1) transgenic mice, in which the orexin gene is knocked out, showed a marked decrease in the amount of Aß pathology in the brain with an increase in sleep time. Focal overexpression of orexin in the hippocampus in APP/PS1 mice did not alter the total amount of sleep/wakefulness and the amount of Aß pathology. In contrast, sleep deprivation or increasing wakefulness by rescue of orexinergic neurons in APP/PS1 mice lacking orexin increased the amount of Aß pathology in the brain. Collectively, modulation of orexin and its effects on sleep appear to modulate Aß pathology in the brain.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , Sono/fisiologia , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Doença Crônica , Ritmo Circadiano/fisiologia , Vetores Genéticos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Camundongos Knockout , Neuropeptídeos/deficiência , Orexinas , Presenilina-1/metabolismo , Regiões Promotoras Genéticas/genética , Privação do Sono/complicações , Privação do Sono/patologia , Privação do Sono/fisiopatologia , Vigília/fisiologia
6.
Mol Neurodegener ; 9: 20, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24893973

RESUMO

BACKGROUND: Recent genome-wide association studies linked variants in TREM2 to a strong increase in the odds of developing Alzheimer's disease. The mechanism by which TREM2 influences the susceptibility to Alzheimer's disease is currently unknown. TREM2 is expressed by microglia and is thought to regulate phagocytic and inflammatory microglial responses to brain pathology. Given that a single allele of variant TREM2, likely resulting in a loss of function, conferred an increased risk of developing Alzheimer's disease, we tested whether loss of one functional trem2 allele would affect Aß plaque deposition or the microglial response to Aß pathology in APPPS1-21 mice. RESULTS: There was no significant difference in Aß deposition in 3-month old or 7-month old APPPS1-21 mice expressing one or two copies of trem2. However, 3-month old mice with one copy of trem2 exhibited a marked decrease in the number and size of plaque-associated microglia. While there were no statistically significant differences in cytokine levels or markers of microglial activation in 3- or 7-month old animals, there were trends towards decreased expression of NOS2, C1qa, and IL1a in 3-month old TREM2+/- vs. TREM2+/+ mice. CONCLUSIONS: Loss of a single copy of trem2 had no effect on Aß pathology, but altered the morphological phenotype of plaque-associated microglia. These data suggest that TREM2 is important for the microglial response to Aß deposition but that a 50% decrease inTREM2 expression does not affect Aß plaque burden.


Assuntos
Doença de Alzheimer , Glicoproteínas de Membrana/genética , Microglia/metabolismo , Placa Amiloide/genética , Receptores Imunológicos/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Feminino , Heterozigoto , Masculino , Camundongos , Camundongos Transgênicos , Microglia/patologia , Placa Amiloide/patologia , Presenilina-1/genética , Reação em Cadeia da Polimerase em Tempo Real
7.
J Exp Med ; 211(3): 387-93, 2014 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-24534188

RESUMO

Tau is primarily a cytoplasmic protein that stabilizes microtubules. However, it is also found in the extracellular space of the brain at appreciable concentrations. Although its presence there may be relevant to the intercellular spread of tau pathology, the cellular mechanisms regulating tau release into the extracellular space are not well understood. To test this in the context of neuronal networks in vivo, we used in vivo microdialysis. Increasing neuronal activity rapidly increased the steady-state levels of extracellular tau in vivo. Importantly, presynaptic glutamate release is sufficient to drive tau release. Although tau release occurred within hours in response to neuronal activity, the elimination rate of tau from the extracellular compartment and the brain is slow (half-life of ∼11 d). The in vivo results provide one mechanism underlying neuronal tau release and may link trans-synaptic spread of tau pathology with synaptic activity itself.


Assuntos
Encéfalo/metabolismo , Espaço Extracelular/metabolismo , Neurônios/metabolismo , Transmissão Sináptica/fisiologia , Tauopatias/fisiopatologia , Proteínas tau/metabolismo , Análise de Variância , Animais , Eletroencefalografia , Ensaio de Imunoadsorção Enzimática , Feminino , Ácido Glutâmico/metabolismo , Meia-Vida , Cinética , Luciferases , Masculino , Camundongos , Camundongos Transgênicos , Microdiálise , Tetrodotoxina
8.
J Neurosci ; 33(31): 12887-97, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23904623

RESUMO

Tau, a microtubule-associated protein, is implicated in the pathogenesis of Alzheimer's Disease (AD) in regard to both neurofibrillary tangle formation and neuronal network hyperexcitability. The genetic ablation of tau substantially reduces hyperexcitability in AD mouse lines, induced seizure models, and genetic in vivo models of epilepsy. These data demonstrate that tau is an important regulator of network excitability. However, developmental compensation in the genetic tau knock-out line may account for the protective effect against seizures. To test the efficacy of a tau reducing therapy for disorders with a detrimental hyperexcitability profile in adult animals, we identified antisense oligonucleotides that selectively decrease endogenous tau expression throughout the entire mouse CNS--brain and spinal cord tissue, interstitial fluid, and CSF--while having no effect on baseline motor or cognitive behavior. In two chemically induced seizure models, mice with reduced tau protein had less severe seizures than control mice. Total tau protein levels and seizure severity were highly correlated, such that those mice with the most severe seizures also had the highest levels of tau. Our results demonstrate that endogenous tau is integral for regulating neuronal hyperexcitability in adult animals and suggest that an antisense oligonucleotide reduction of tau could benefit those with epilepsy and perhaps other disorders associated with tau-mediated neuronal hyperexcitability.


Assuntos
Anticonvulsivantes/uso terapêutico , Oligonucleotídeos Antissenso/uso terapêutico , Convulsões/prevenção & controle , Proteínas tau/genética , Fatores Etários , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Convulsivantes/toxicidade , Modelos Animais de Doenças , Transtornos Neurológicos da Marcha/tratamento farmacológico , Transtornos Neurológicos da Marcha/etiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Infusões Intraventriculares , Ácido Láctico/metabolismo , Locomoção/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Pentilenotetrazol/toxicidade , Picrotoxina/toxicidade , Convulsões/induzido quimicamente , Convulsões/genética , Proteínas tau/química , Proteínas tau/metabolismo
9.
J Exp Med ; 209(12): 2149-56, 2012 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-23129750

RESUMO

The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for Alzheimer's disease (AD). The influence of apoE on amyloid ß (Aß) accumulation may be the major mechanism by which apoE affects AD. ApoE interacts with Aß and facilitates Aß fibrillogenesis in vitro. In addition, apoE is one of the protein components in plaques. We hypothesized that certain anti-apoE antibodies, similar to certain anti-Aß antibodies, may have antiamyloidogenic effects by binding to apoE in the plaques and activating microglia-mediated amyloid clearance. To test this hypothesis, we developed several monoclonal anti-apoE antibodies. Among them, we administered HJ6.3 antibody intraperitoneally to 4-mo-old male APPswe/PS1ΔE9 mice weekly for 14 wk. HJ6.3 dramatically decreased amyloid deposition by 60-80% and significantly reduced insoluble Aß40 and Aß42 levels. Short-term treatment with HJ6.3 resulted in strong changes in microglial responses around Aß plaques. Collectively, these results suggest that anti-apoE immunization may represent a novel AD therapeutic strategy and that other proteins involved in Aß binding and aggregation might also be a target for immunotherapy. Our data also have important broader implications for other amyloidosis. Immunotherapy to proteins tightly associated with misfolded proteins might open up a new treatment option for many protein misfolding diseases.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloidose/prevenção & controle , Anticorpos Monoclonais/farmacologia , Apolipoproteínas E/imunologia , Encefalopatias/prevenção & controle , Imunoterapia/métodos , Amiloidose/imunologia , Amiloidose/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/sangue , Apolipoproteínas E/metabolismo , Encefalopatias/imunologia , Encefalopatias/metabolismo , Ensaio de Imunoadsorção Enzimática , Immunoblotting , Imunoprecipitação , Masculino , Camundongos , Camundongos Transgênicos , Estatísticas não Paramétricas
10.
Int J Clin Exp Pathol ; 5(6): 482-95, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22949930

RESUMO

Alzheimer's disease is characterized in part by extracellular aggregation of the amyloid-ß peptide in the form of diffuse and fibrillar plaques in the brain. Electron microscopy (EM) has made an important contribution in understanding of the structure of amyloid plaques in humans. Classical EM studies have revealed the architecture of the fibrillar core, characterized the progression of neuritic changes, and have identified the neurofibrillary tangles formed by paired helical filaments (PHF) in degenerating neurons. Clinical data has strongly correlated cognitive impairment in AD with the substantial synapse loss observed in these early ultrastructural studies. Animal models of AD-type brain amyloidosis have provided excellent opportunities to study amyloid and neuritic pathology in detail and establish the role of neurons and glia in plaque formation. Transgenic mice overexpressing mutant amyloid precursor protein (APP) alone with or without mutant presenilin 1 (PS1), have shown that brain amyloid plaque development and structure grossly recapitulate classical findings in humans. Transgenic APP/PS1 mice expressing human apolioprotein E isoforms also develop amyloid plaque deposition. However no ultrastructural data has been reported for these animals. Here we show results from detailed EM analysis of amyloid plaques in APP/PS1 mice expressing human isoforms of ApoE and compare these findings with EM data in other transgenic models and in human AD. Our results show that similar to other transgenic animals, APP/PS1 mice expressing human ApoE isoforms share all major cellular and subcellular degenerative features and highlight the identity of the cellular elements involved in Aß deposition and neuronal degeneration.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Apolipoproteínas E/metabolismo , Placa Amiloide/ultraestrutura , Presenilina-1/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/ultraestrutura , Amiloidose/patologia , Animais , Apolipoproteínas E/ultraestrutura , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação , Neurônios/metabolismo , Neurônios/ultraestrutura , Placa Amiloide/metabolismo , Presenilina-1/ultraestrutura , Isoformas de Proteínas
11.
Sci Transl Med ; 4(150): 150ra122, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956200

RESUMO

Aggregation of ß-amyloid (Aß) in the brain begins to occur years before the clinical onset of Alzheimer's disease (AD). Before Aß aggregation, concentrations of extracellular soluble Aß in the interstitial fluid (ISF) space of the brain, which are regulated by neuronal activity and the sleep-wake cycle, correlate with the amount of Aß deposition in the brain seen later. The amount and quality of sleep decline with normal aging and to a greater extent in AD patients. How sleep quality as well as the diurnal fluctuation in Aß change with age and Aß aggregation is not well understood. We report a normal sleep-wake cycle and diurnal fluctuation in ISF Aß in the brain of the APPswe/PS1δE9 mouse model of AD before Aß plaque formation. After plaque formation, the sleep-wake cycle markedly deteriorated and diurnal fluctuation of ISF Aß dissipated. As in mice, diurnal fluctuation of cerebrospinal fluid Aß in young adult humans with presenilin mutations was also markedly attenuated after Aß plaque formation. Virtual elimination of Aß deposits in the mouse brain by active immunization with Aß(42) normalized the sleep-wake cycle and the diurnal fluctuation of ISF Aß. These data suggest that Aß aggregation disrupts the sleep-wake cycle and diurnal fluctuation of Aß. Sleep-wake behavior and diurnal fluctuation of Aß in the central nervous system may be functional and biochemical indicators, respectively, of Aß-associated pathology.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Ritmo Circadiano/fisiologia , Sono/fisiologia , Vigília/fisiologia , Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Animais , Modelos Animais de Doenças , Líquido Extracelular/metabolismo , Genes Dominantes/genética , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imunização , Lactatos/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Neostriado/metabolismo , Neostriado/patologia , Presenilinas/genética , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 109(38): 15502-7, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22927427

RESUMO

The apolipoprotein E (APOE)-ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer's disease, likely increasing risk by altering amyloid-ß (Aß) accumulation. We recently demonstrated that the low-density lipoprotein receptor (LDLR) is a major apoE receptor in the brain that strongly regulates amyloid plaque deposition. In the current study, we sought to understand the mechanism by which LDLR regulates Aß accumulation by altering Aß clearance from brain interstitial fluid. We hypothesized that increasing LDLR levels enhances blood-brain barrier-mediated Aß clearance, thus leading to reduced Aß accumulation. Using the brain Aß efflux index method, we found that blood-brain barrier-mediated clearance of exogenously administered Aß is enhanced with LDLR overexpression. We next developed a method to directly assess the elimination of centrally derived, endogenous Aß into the plasma of mice using an anti-Aß antibody that prevents degradation of plasma Aß, allowing its rate of appearance from the brain to be measured. Using this plasma Aß accumulation technique, we found that LDLR overexpression enhances brain-to-blood Aß transport. Together, our results suggest a unique mechanism by which LDLR regulates brain-to-blood Aß clearance, which may serve as a useful therapeutic avenue in targeting Aß clearance from the brain.


Assuntos
Amiloidose/metabolismo , Apolipoproteína E4/genética , Receptores de LDL/biossíntese , Alelos , Peptídeos beta-Amiloides/metabolismo , Animais , Barreira Hematoencefálica , Encéfalo/metabolismo , Modelos Animais de Doenças , Insulina/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Microdiálise , Transgenes
13.
J Neurosci ; 32(13): 4334-40, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22457485

RESUMO

Brain region-specific deposition of extracellular amyloid plaques principally composed of aggregated amyloid-ß (Aß) peptide is a pathological signature of Alzheimer's disease (AD). Recent human neuroimaging data suggest that resting-state functional connectivity strength is reduced in patients with AD, cognitively normal elderly harboring elevated amyloid burden, and in advanced aging. Interestingly, there exists a striking spatial correlation between functional connectivity strength in cognitively normal adults and the location of Aß plaque deposition in AD. However, technical limitations have heretofore precluded examination of the relationship between functional connectivity, Aß deposition, and normal aging in mouse models. Using a novel functional connectivity optical intrinsic signal (fcOIS) imaging technique, we demonstrate that Aß deposition is associated with significantly reduced bilateral functional connectivity in multiple brain regions of older APP/PS1 transgenic mice. The amount of Aß deposition in each brain region was associated with the degree of local, age-related bilateral functional connectivity decline. Normal aging was associated with reduced bilateral functional connectivity specifically in retrosplenial cortex. Furthermore, we found that the magnitude of regional bilateral functional correlation in young APP/PS1 mice before Aß plaque formation was proportional to the amount of region-specific plaque deposition seen later in older APP/PS1 mice. Together, these findings suggest that Aß deposition and normal aging are associated with region-specific disruption of functional connectivity and that the magnitude of local bilateral functional connectivity predicts regional vulnerability to subsequent Aß deposition in mouse brain.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/fisiopatologia , Neuroimagem Funcional/estatística & dados numéricos , Vias Neurais/fisiopatologia , Placa Amiloide/metabolismo , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Amiloidose/metabolismo , Amiloidose/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Neuroimagem Funcional/métodos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Vias Neurais/metabolismo
14.
J Neurosci ; 31(49): 18007-12, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22159114

RESUMO

The ε4 allele of the apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease (AD). Evidence suggests that the effect of apoE isoforms on amyloid-ß (Aß) accumulation in the brain plays a critical role in AD pathogenesis. Like in humans, apoE4 expression in animal models that develop Aß amyloidosis results in greater Aß and amyloid deposition than with apoE3 expression. However, whether decreasing levels of apoE3 or apoE4 would promote or attenuate Aß-related pathology has not been directly addressed. To determine the effect of decreasing human apoE levels on Aß accumulation in vivo, we generated human APOE isoform haploinsufficient mouse models by crossing APPPS1-21 mice with APOE isoform knock-in mice. By genetically manipulating APOE gene dosage, we demonstrate that decreasing human apoE levels, regardless of isoform status, results in significantly decreased amyloid plaque deposition and microglial activation. These differences in amyloid load between apoE3- and apoE4-expressing mice were not due to apoE4 protein being present at lower levels than apoE3. These data suggest that current therapeutic strategies to increase apoE levels without altering its lipidation state may actually worsen Aß amyloidosis, while increasing apoE degradation or inhibiting its synthesis may be a more effective treatment approach.


Assuntos
Amiloide/metabolismo , Amiloidose/genética , Amiloidose/metabolismo , Apolipoproteínas E/deficiência , Haploinsuficiência/genética , Precursor de Proteína beta-Amiloide/genética , Amiloidose/patologia , Animais , Apolipoproteínas E/genética , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Placa Amiloide/patologia , Presenilina-1/genética , Isoformas de Proteínas/genética
15.
J Neurosci ; 31(37): 13110-7, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917794

RESUMO

Although tau is a cytoplasmic protein, it is also found in brain extracellular fluids, e.g., CSF. Recent findings suggest that aggregated tau can be transferred between cells and extracellular tau aggregates might mediate spread of tau pathology. Despite these data, details of whether tau is normally released into the brain interstitial fluid (ISF), its concentration in ISF in relation to CSF, and whether ISF tau is influenced by its aggregation are unknown. To address these issues, we developed a microdialysis technique to analyze monomeric ISF tau levels within the hippocampus of awake, freely moving mice. We detected tau in ISF of wild-type mice, suggesting that tau is released in the absence of neurodegeneration. ISF tau was significantly higher than CSF tau and their concentrations were not significantly correlated. Using P301S human tau transgenic mice (P301S tg mice), we found that ISF tau is fivefold higher than endogenous murine tau, consistent with its elevated levels of expression. However, following the onset of tau aggregation, monomeric ISF tau decreased markedly. Biochemical analysis demonstrated that soluble tau in brain homogenates decreased along with the deposition of insoluble tau. Tau fibrils injected into the hippocampus decreased ISF tau, suggesting that extracellular tau is in equilibrium with extracellular or intracellular tau aggregates. This technique should facilitate further studies of tau secretion, spread of tau pathology, the effects of different disease states on ISF tau, and the efficacy of experimental treatments.


Assuntos
Envelhecimento/metabolismo , Líquido Extracelular/metabolismo , Hipocampo/metabolismo , Microdiálise/métodos , Proteínas tau/genética , Proteínas tau/metabolismo , Envelhecimento/líquido cefalorraquidiano , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Neurológicos , Solubilidade , Proteínas tau/administração & dosagem , Proteínas tau/líquido cefalorraquidiano , Proteínas tau/química
16.
Sci Transl Med ; 3(89): 89ra57, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21715678

RESUMO

The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer's disease (AD). The APOE ε4 allele markedly increases AD risk and decreases age of onset, likely through its strong effect on the accumulation of amyloid-ß (Aß) peptide. In contrast, the APOE ε2 allele appears to decrease AD risk. Most rare, early-onset forms of familial AD are caused by autosomal dominant mutations that often lead to overproduction of Aß(42) peptide. However, the mechanism by which APOE alleles differentially modulate Aß accumulation in sporadic, late-onset AD is less clear. In a cohort of cognitively normal individuals, we report that reliable molecular and neuroimaging biomarkers of cerebral Aß deposition vary in an apoE isoform-dependent manner. We hypothesized that human apoE isoforms differentially affect Aß clearance or synthesis in vivo, resulting in an apoE isoform-dependent pattern of Aß accumulation later in life. Performing in vivo microdialysis in a mouse model of Aß-amyloidosis expressing human apoE isoforms (PDAPP/TRE), we find that the concentration and clearance of soluble Aß in the brain interstitial fluid depends on the isoform of apoE expressed. This pattern parallels the extent of Aß deposition observed in aged PDAPP/TRE mice. ApoE isoform-dependent differences in soluble Aß metabolism are observed not only in aged but also in young PDAPP/TRE mice well before the onset of Aß deposition in amyloid plaques in the brain. Additionally, amyloidogenic processing of amyloid precursor protein and Aß synthesis, as assessed by in vivo stable isotopic labeling kinetics, do not vary according to apoE isoform in young PDAPP/TRE mice. Our results suggest that APOE alleles contribute to AD risk by differentially regulating clearance of Aß from the brain, suggesting that Aß clearance pathways may be useful therapeutic targets for AD prevention.


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Isoformas de Proteínas/metabolismo , Adulto , Idoso , Alelos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Apolipoproteína E4/genética , Biomarcadores/líquido cefalorraquidiano , Encéfalo/patologia , Feminino , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microdiálise , Pessoa de Meia-Idade , Isoformas de Proteínas/genética
17.
Nat Neurosci ; 14(6): 750-6, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21532579

RESUMO

Amyloid-ß (Aß) plaque deposition in specific brain regions is a pathological hallmark of Alzheimer's disease. However, the mechanism underlying the regional vulnerability to Aß deposition in Alzheimer's disease is unknown. Herein, we provide evidence that endogenous neuronal activity regulates the regional concentration of interstitial fluid (ISF) Aß, which drives local Aß aggregation. Using in vivo microdialysis, we show that ISF Aß concentrations in several brain regions of APP transgenic mice before plaque deposition were commensurate with the degree of subsequent plaque deposition and with the concentration of lactate, a marker of neuronal activity. Furthermore, unilateral vibrissal stimulation increased ISF Aß, and unilateral vibrissal deprivation decreased ISF Aß and lactate, in contralateral barrel cortex. Long-term unilateral vibrissal deprivation decreased amyloid plaque formation and growth. Our results suggest a mechanism to account for the vulnerability of specific brain regions to Aß deposition in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/patologia , Neurônios/metabolismo , Privação Sensorial , Doença de Alzheimer , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Líquido Extracelular , Feminino , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microdiálise , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Córtex Somatossensorial/patologia
18.
Neuron ; 58(1): 42-51, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18400162

RESUMO

Aggregation of amyloid-beta (Abeta) peptide into soluble and insoluble forms within the brain extracellular space is central to the pathogenesis of Alzheimer's disease. Full-length amyloid precursor protein (APP) is endocytosed from the cell surface into endosomes where it is cleaved to produce Abeta. Abeta is subsequently released into the brain interstitial fluid (ISF). We hypothesized that synaptic transmission results in more APP endocytosis, thereby increasing Abeta generation and release into the ISF. We found that inhibition of clathrin-mediated endocytosis immediately lowers ISF Abeta levels in vivo. Two distinct methods that increased synaptic transmission resulted in an elevation of ISF Abeta levels. Inhibition of endocytosis, however, prevented the activity-dependent increase in Abeta. We estimate that approximately 70% of ISF Abeta arises from endocytosis-associated mechanisms, with the vast majority of this pool also dependent on synaptic activity. These findings have implications for AD pathogenesis and may provide insights into therapeutic intervention.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Endocitose , Sinapses/metabolismo , Transmissão Sináptica , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/fisiologia , Animais , Endocitose/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sinapses/genética , Transmissão Sináptica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...