Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 194: 106473, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38493903

RESUMO

The pathophysiological process of Alzheimer's disease (AD) is believed to begin many years before the formal diagnosis of AD dementia. This protracted preclinical phase offers a crucial window for potential therapeutic interventions, yet its comprehensive characterization remains elusive. Accumulating evidence suggests that amyloid-ß (Aß) may mediate neuronal hyperactivity in circuit dysfunction in the early stages of AD. At the same time, neural activity can also facilitate Aß accumulation through intricate feed-forward interactions, complicating elucidating the conditions governing Aß-dependent hyperactivity and its diagnostic utility. In this study, we use biophysical modeling to shed light on such conditions. Our analysis reveals that the inherently nonlinear nature of the underlying molecular interactions can give rise to the emergence of various modes of hyperactivity. This diversity in the mechanisms of hyperactivity may ultimately account for a spectrum of AD manifestations.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Neurônios/fisiologia , Comunicação Celular
2.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38731812

RESUMO

We compared the clinical and analytical performance of Alzheimer's disease (AD) plasma biomarkers measured using the single-molecule array (Simoa) and Lumipulse platforms. We quantified the plasma levels of amyloid beta 42 (Aß42), Aß40, phosphorylated tau (Ptau181), and total tau biomarkers in 81 patients with mild cognitive impairment (MCI), 30 with AD, and 16 with non-AD dementia. We found a strong correlation between the Simoa and Lumipulse methods. Concerning the clinical diagnosis, Simoa Ptau181/Aß42 (AUC 0.739, 95% CI 0.592-0.887) and Lumipulse Aß42 and Ptau181/Aß42 (AUC 0.735, 95% CI 0.589-0.882 and AUC 0.733, 95% CI 0.567-0.900) had the highest discriminating power. However, their power was significantly lower than that of CSF Aß42/Aß40, as measured by Lumipulse (AUC 0.879, 95% CI 0.766-0.992). Simoa Ptau181 and Lumipulse Ptau181/Aß42 were the markers most consistent with the CSF Aß42/Aß40 status (AUC 0.801, 95% CI 0.712-0.890 vs. AUC 0.870, 95% CI 0.806-0.934, respectively) at the ≥2.127 and ≥0.084 cut-offs, respectively. The performance of the Simoa and Lumipulse plasma AD assays is weaker than that of CSF AD biomarkers. At present, the analysed AD plasma biomarkers may be useful for screening to reduce the number of lumbar punctures in the clinical setting.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Biomarcadores , Disfunção Cognitiva , Proteínas tau , Humanos , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/sangue , Doença de Alzheimer/diagnóstico , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Masculino , Feminino , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/sangue , Idoso , Proteínas tau/líquido cefalorraquidiano , Proteínas tau/sangue , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/sangue , Pessoa de Meia-Idade , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fragmentos de Peptídeos/sangue , Idoso de 80 Anos ou mais , Fosforilação
3.
Int J Mol Sci ; 23(10)2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35628557

RESUMO

Glial cells participate actively in the early cognitive decline in Alzheimer's disease (AD) pathology. In fact, recent studies have found molecular and functional abnormalities in astrocytes and microglia in both animal models and brains of patients suffering from this pathology. In this regard, reactive gliosis intimately associated with amyloid plaques has become a pathological hallmark of AD. A recent study from our laboratory reports that astrocyte reactivity is caused by a direct interaction between amyloid beta (Aß) oligomers and integrin ß1. Here, we have generated four recombinant peptides including the extracellular domain of integrin ß1, and evaluated their capacity both to bind in vitro to Aß oligomers and to prevent in vivo Aß oligomer-induced gliosis and endoplasmic reticulum stress. We have identified the minimal region of integrin ß1 that binds to Aß oligomers. This region is called signal peptide and corresponds to the first 20 amino acids of the integrin ß1 N-terminal domain. This recombinant integrin ß1 signal peptide prevented Aß oligomer-induced ROS generation in primary astrocyte cultures. Furthermore, we carried out intrahippocampal injection in adult mice of recombinant integrin ß1 signal peptide combined with or without Aß oligomers and we evaluated by immunohistochemistry both astrogliosis and microgliosis as well as endoplasmic reticulum stress. The results show that recombinant integrin ß1 signal peptide precluded both astrogliosis and microgliosis and endoplasmic reticulum stress mediated by Aß oligomers in vivo. We have developed a molecular tool that blocks the activation of the molecular cascade that mediates gliosis via Aß oligomer/integrin ß1 signaling.


Assuntos
Peptídeos beta-Amiloides , Gliose , Integrina beta1 , Sinais Direcionadores de Proteínas , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Integrina beta1/metabolismo , Camundongos
4.
Int J Mol Sci ; 23(16)2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-36012525

RESUMO

We have previously proposed a radical change in the current strategy to clear pathogenic proteins from the central nervous system (CNS) based on the cerebrospinal fluid (CSF)-sink therapeutic strategy, whereby pathogenic proteins can be removed directly from the CNS via CSF. To this aim, we designed and manufactured an implantable device for selective and continuous apheresis of CSF enabling, in combination with anti-amyloid-beta (Aß) monoclonal antibodies (mAb), the clearance of Aß from the CSF. Here, we provide the first proof of concept in the APP/PS1 mouse model of Alzheimer's disease (AD). Devices were implanted in twenty-four mice (seventeen APP/PS1 and seven Wt) with low rates of complications. We confirmed that the apheresis module is permeable to the Aß peptide and impermeable to mAb. Moreover, our results showed that continuous clearance of soluble Aß from the CSF for a few weeks decreases cortical Aß plaques. Thus, we conclude that this intervention is feasible and may provide important advantages in terms of safety and efficacy.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo
5.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572157

RESUMO

Platelet-rich plasma (PRP) is a biologic therapy that promotes healing responses across multiple medical fields, including the central nervous system (CNS). The efficacy of this therapy depends on several factors such as the donor's health status and age. This work aims to prove the effect of PRP on cellular models of the CNS, considering the differences between PRP from young and elderly donors. Two different PRP pools were prepared from donors 65‒85 and 20‒25 years old. The cellular and molecular composition of both PRPs were analyzed. Subsequently, the cellular response was evaluated in CNS in vitro models, studying proliferation, neurogenesis, synaptogenesis, and inflammation. While no differences in the cellular composition of PRPs were found, the molecular composition of the Young PRP showed lower levels of inflammatory molecules such as CCL-11, as well as the presence of other factors not found in Aged PRP (GDF-11). Although both PRPs had effects in terms of reducing neural progenitor cell apoptosis, stabilizing neuronal synapses, and decreasing inflammation in the microglia, the effect of the Young PRP was more pronounced. In conclusion, the molecular composition of the PRP, conditioned by the age of the donors, affects the magnitude of the biological response.


Assuntos
Córtex Cerebral/imunologia , Mediadores da Inflamação/metabolismo , Microglia/imunologia , Plasma Rico em Plaquetas/imunologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/imunologia , Animais , Apoptose/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Córtex Cerebral/citologia , Quimiocina CCL11/metabolismo , Feminino , Humanos , Masculino , Camundongos , Microglia/citologia , Células-Tronco Neurais/imunologia , Neurogênese/imunologia , Neurônios/imunologia , Plasma Rico em Plaquetas/citologia , Plasma Rico em Plaquetas/metabolismo , Cultura Primária de Células , Ratos , Sinapses/imunologia , Adulto Jovem
6.
Mol Cell Neurosci ; 95: 86-95, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30763689

RESUMO

The normal role of Alzheimer's disease (AD)-linked amyloid precursor protein (APP) in the brain remains incompletely understood. Previous studies have reported that lack of APP has detrimental effects on spines and electrophysiological parameters. APP has been described to be important in synaptic pruning during development. The effect of APP knockout on mature synapses is complicated by this role in development. We previously reported on differential changes in synaptic proteins and receptors in APP mutant AD transgenic compared to wild-type neurons, which revealed selective decreases in levels of pre- and post-synaptic proteins, including of surface glutamate receptors. In the present study, we undertook a similar analysis of synaptic composition but now in APP knockout compared to wild-type mouse neurons. Here we demonstrate alterations in levels of selective pre- and post-synaptic proteins and receptors in APP knockout compared to wild-type mouse primary neurons in culture and brains of mice in youth and adulthood. Remarkably, we demonstrate selective increases in levels of synaptic proteins, such as GluA1, in neurons with APP knockout and with RNAi knockdown, which tended to be opposite to the reductions seen in AD transgenic APP mutant compared to wild-type neurons. These data reinforce that APP is important for the normal composition of synapses.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Neurônios/metabolismo , Sinapses/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Proteína 4 Homóloga a Disks-Large/genética , Proteína 4 Homóloga a Disks-Large/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo
8.
Mediators Inflamm ; 2018: 2530414, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30533998

RESUMO

Synapse loss is an early manifestation of pathology in Alzheimer's disease (AD) and is currently the best correlate to cognitive decline. Microglial cells are involved in synapse pruning during development via the complement pathway. Moreover, recent evidence points towards a key role played by glial cells in synapse loss during AD. However, further contribution of glial cells and the role of neurons to synapse pathology in AD remain not well understood. This review is aimed at comprehensively reporting the source and/or cellular localization in the CNS-in microglia, astrocytes, or neurons-of the triggering components (C1q, C3) of the classical complement pathway involved in synapse pruning in development, adulthood, and AD.


Assuntos
Envelhecimento/fisiologia , Doença de Alzheimer/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Humanos , Microglia/metabolismo , Neurônios/citologia
9.
Arterioscler Thromb Vasc Biol ; 35(10): 2092-103, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26293468

RESUMO

OBJECTIVE: Although dendritic cells are known to play a role in atherosclerosis, few studies have examined the contribution of the wide variety of dendritic cell subsets. Accordingly, their roles in atherogenesis remain largely unknown. We investigated the ability of different dendritic cell subsets to become foam cells after contact with aggregated low-density lipoprotein (LDL; the predominant form of LDL found in atherosclerotic plaques). APPROACH AND RESULTS: We demonstrate that both murine and human monocyte-derived dendritic cells use exophagy to degrade aggregated LDL, leading to foam cell formation, whereas monocyte-independent dendritic cells are unable to clear LDL aggregates by this mechanism. Exophagy is a catabolic process in which objects that cannot be internalized by phagocytosis (because of their size or association with extracellular structures) are initially digested in an extracellular acidic lytic compartment. Surprisingly, we found that monocyte-derived dendritic cells upregulate exophagy on maturation. This contrasts various forms of endocytic internalization in dendritic cells, which decrease on maturation. Finally, we show that our in vitro results are consistent with dendritic cell lipid accumulation in plaques of an ApoE(-/-) mouse model of atherosclerosis. CONCLUSIONS: Our results show that monocyte-derived dendritic cells use exophagy to degrade aggregated LDL and become foam cells, whereas monocyte-independent dendritic cells are unable to clear LDL deposits. Furthermore, we find that exophagy is upregulated on dendritic cell maturation. Thus, exophagy-mediated foam cell formation in monocyte-derived dendritic cells could play a significant role in atherogenesis.


Assuntos
Aterosclerose/genética , Aterosclerose/patologia , Células Dendríticas/citologia , Células Espumosas/citologia , Lipoproteínas LDL/metabolismo , Fagocitose/fisiologia , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Células Espumosas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Monócitos/citologia , Monócitos/metabolismo , Fagocitose/genética , Distribuição Aleatória , Ativação Transcricional , Regulação para Cima
10.
J Neurosci ; 32(4): 1273-83, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22279212

RESUMO

An early role of amyloid-ß peptide (Aß) aggregation in Alzheimer's disease pathogenesis is well established. However, the contribution of intracellular or extracellular forms of Aß to the neurodegenerative process is a subject of considerable debate. We here describe transgenic mice expressing Aß1-40 (APP47) and Aß1-42 (APP48) with a cleaved signal sequence to insert both peptides during synthesis into the endoplasmic reticulum. Although lower in transgene mRNA, APP48 mice reach a higher brain Aß concentration. The reduced solubility and increased aggregation of Aß1-42 may impair its degradation. APP48 mice develop intracellular Aß lesions in dendrites and lysosomes. The hippocampal neuron number is reduced already at young age. The brain weight decreases during aging in conjunction with severe white matter atrophy. The mice show a motor impairment. Only very few Aß1-40 lesions are found in APP47 mice. Neither APP47 nor APP48 nor the bigenic mice develop extracellular amyloid plaques. While intracellular membrane expression of Aß1-42 in APP48 mice does not lead to the AD-typical lesions, Aß aggregates develop within cells accompanied by considerable neurodegeneration.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Regulação da Expressão Gênica , Degeneração Neural/genética , Neurônios/metabolismo , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/patologia , Ratos
11.
Autophagy ; 19(2): 660-677, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35867714

RESUMO

Synapses represent an important target of Alzheimer disease (AD), and alterations of their excitability are among the earliest changes associated with AD development. Synaptic activation has been shown to be protective in models of AD, and deep brain stimulation (DBS), a surgical strategy that modulates neuronal activity to treat neurological and psychiatric disorders, produced positive effects in AD patients. However, the molecular mechanisms underlying the protective role(s) of brain stimulation are still elusive. We have previously demonstrated that induction of synaptic activity exerts protection in mouse models of AD and frontotemporal dementia (FTD) by enhancing the macroautophagy/autophagy flux and lysosomal degradation of pathological MAPT/Tau. We now provide evidence that TFEB (transcription factor EB), a master regulator of lysosomal biogenesis and autophagy, is a key mediator of this cellular response. In cultured primary neurons from FTD-transgenic mice, synaptic stimulation inhibits MTORC1 signaling, thus promoting nuclear translocation of TFEB, which, in turn, induces clearance of MAPT/Tau oligomers. Conversely, synaptic activation fails to promote clearance of toxic MAPT/Tau in neurons expressing constitutively active RRAG GTPases, which sequester TFEB in the cytosol, or upon TFEB depletion. Activation of TFEB is also confirmed in vivo in DBS-stimulated AD mice. We also demonstrate that DBS reduces pathological MAPT/Tau and promotes neuroprotection in Parkinson disease patients with tauopathy. Altogether our findings indicate that stimulation of synaptic activity promotes TFEB-mediated clearance of pathological MAPT/Tau. This mechanism, underlying the protective effect of DBS, provides encouraging support for the use of synaptic stimulation as a therapeutic treatment against tauopathies.Abbreviations: 3xTg-AD: triple transgenic AD mice; AD: Alzheimer disease; CSA: cyclosporine A; DBS: deep brain stimulation; DIV: days in vitro; EC: entorhinal cortex; FTD: frontotemporal dementia; gLTP: glycine-induced long-term potentiation; GPi: internal segment of the globus pallidus; PD: Parkinson disease; STN: subthalamic nucleus; TFEB: transcription factor EB.


Assuntos
Doença de Alzheimer , Demência Frontotemporal , Doença de Parkinson , Tauopatias , Camundongos , Animais , Doença de Alzheimer/metabolismo , Demência Frontotemporal/metabolismo , Doença de Parkinson/metabolismo , Autofagia , Tauopatias/metabolismo , Camundongos Transgênicos , Lisossomos/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proteínas tau/metabolismo
12.
Autophagy ; 19(7): 1952-1981, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36622892

RESUMO

Microglial phagocytosis of apoptotic debris prevents buildup damage of neighbor neurons and inflammatory responses. Whereas microglia are very competent phagocytes under physiological conditions, we report their dysfunction in mouse and preclinical monkey models of stroke (macaques and marmosets) by transient occlusion of the medial cerebral artery (tMCAo). By analyzing recently published bulk and single cell RNA sequencing databases, we show that the phagocytosis dysfunction was not explained by transcriptional changes. In contrast, we demonstrate that the impairment of both engulfment and degradation was related to energy depletion triggered by oxygen and nutrient deprivation (OND), which led to reduced process motility, lysosomal exhaustion, and the induction of a protective macroautophagy/autophagy response in microglia. Basal autophagy, in charge of removing and recycling intracellular elements, was critical to maintain microglial physiology, including survival and phagocytosis, as we determined both in vivo and in vitro using pharmacological and transgenic approaches. Notably, the autophagy inducer rapamycin partially prevented the phagocytosis impairment induced by tMCAo in vivo but not by OND in vitro, where it even had a detrimental effect on microglia, suggesting that modulating microglial autophagy to optimal levels may be a hard to achieve goal. Nonetheless, our results show that pharmacological interventions, acting directly on microglia or indirectly on the brain environment, have the potential to recover phagocytosis efficiency in the diseased brain. We propose that phagocytosis is a therapeutic target yet to be explored in stroke and other brain disorders and provide evidence that it can be modulated in vivo using rapamycin.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; AMBRA1: autophagy/beclin 1 regulator 1; ATG4B: autophagy related 4B, cysteine peptidase; ATP: adenosine triphosphate; BECN1: beclin 1, autophagy related; CASP3: caspase 3; CBF: cerebral blood flow; CCA: common carotid artery; CCR2: chemokine (C-C motif) receptor 2; CIR: cranial irradiation; Csf1r/v-fms: colony stimulating factor 1 receptor; CX3CR1: chemokine (C-X3-C motif) receptor 1; DAPI: 4',6-diamidino-2-phenylindole; DG: dentate gyrus; GO: Gene Ontology; HBSS: Hanks' balanced salt solution; HI: hypoxia-ischemia; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MCA: medial cerebral artery; MTOR: mechanistic target of rapamycin kinase; OND: oxygen and nutrient deprivation; Ph/A coupling: phagocytosis-apoptosis coupling; Ph capacity: phagocytic capacity; Ph index: phagocytic index; SQSTM1: sequestosome 1; RNA-Seq: RNA sequencing; TEM: transmission electron microscopy; tMCAo: transient medial cerebral artery occlusion; ULK1: unc-51 like kinase 1.


Assuntos
Autofagia , Acidente Vascular Cerebral , Animais , Camundongos , Autofagia/fisiologia , Microglia/metabolismo , Proteína Beclina-1/metabolismo , Fagocitose/genética , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo , Oxigênio/farmacologia , Sirolimo/farmacologia
13.
J Neurosci ; 31(43): 15384-90, 2011 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22031884

RESUMO

A central question in Alzheimer's disease (AD) research is what role ß-amyloid peptide (Aß) plays in synaptic dysfunction. Synaptic activity increases Aß secretion, potentially inhibiting synapses, but also decreases intraneuronal Aß, protecting synapses. We now show that levels of secreted Aß fall with time in culture in neurons of AD-transgenic mice, but not wild-type mice. Moreover, the ability of synaptic activity to elevate secreted Aß and reduce intraneuronal Aß becomes impaired in AD-transgenic but not wild-type neurons with time in culture. We demonstrate that synaptic activity promotes an increase in the Aß-degrading protease neprilysin at the cell surface and a concomitant increase in colocalization with Aß42. Remarkably, AD-transgenic but not wild-type neurons show reduced levels of neprilysin with time in culture. This impaired ability to secrete Aß and reduce intraneuronal Aß has important implications for the pathogenesis and treatment of AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/patologia , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Glicina/farmacologia , Guanilato Quinases/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Tiorfano/farmacologia
14.
Am J Pathol ; 179(5): 2551-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21925470

RESUMO

ß-Amyloid (Aß) accumulation and aggregation are hallmarks of Alzheimer's disease (AD). High-resolution three-dimensional (HR-3D) volumetric imaging allows for better analysis of fluorescence confocal microscopy and 3D visualization of Aß pathology in brain. Early intraneuronal Aß pathology was studied in AD transgenic mouse brains by HR-3D volumetric imaging. To better visualize and analyze the development of Aß pathology, thioflavin S staining and immunofluorescence using antibodies against Aß, fibrillar Aß, and structural and synaptic neuronal proteins were performed in the brain tissue of Tg19959, wild-type, and Tg19959-YFP mice at different ages. Images obtained by confocal microscopy were reconstructed into three-dimensional volumetric datasets. Such volumetric imaging of CA1 hippocampus of AD transgenic mice showed intraneuronal onset of Aß42 accumulation and fibrillization within cell bodies, neurites, and synapses before plaque formation. Notably, early fibrillar Aß was evident within individual synaptic compartments, where it was associated with abnormal morphology. In dendrites, increasing intraneuronal thioflavin S correlated with decreases in neurofilament marker SMI32. Fibrillar Aß aggregates could be seen piercing the cell membrane. These data support that Aß fibrillization begins within AD vulnerable neurons, leading to disruption of cytoarchitecture and degeneration of spines and neurites. Thus, HR-3D volumetric image analysis allows for better visualization of intraneuronal Aß pathology and provides new insights into plaque formation in AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Região CA1 Hipocampal/patologia , Placa Amiloide/patologia , Sinapses/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Membrana Celular/metabolismo , Progressão da Doença , Feminino , Imageamento Tridimensional , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Neuritos/patologia , Neurônios/patologia , Sinapses/patologia
15.
Neurodegener Dis ; 10(1-4): 56-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22269167

RESUMO

BACKGROUND: ß-Amyloid (Aß) plaques are a pathological hallmark of Alzheimer's disease (AD) and multiple lines of evidence have linked Aß with AD. However, synapse loss is known as the best pathological correlate of cognitive impairment in AD, and intraneuronal Aß accumulation has been shown to precede plaque pathology. The progression of Aß accumulation to synapse loss and plaque formation remains incomplete. The objective is to investigate the progression of intraneuronal Aß accumulation in the brain. METHODS: To visualize and analyze the development of Aß pathology we perform immunohistochemistry and immunofluorescence microscopy using antibodies against different Aß conformations, synaptic proteins and structural neuronal proteins in brain tissue of AD transgenic mouse models. RESULTS: Our results show the intraneuronal onset of Aß42 accumulation in AD mouse brains with aging. We observe an inverse correlation of Aß and amyloid fibrils with structural proteins within neurites. Images reveal aggregated amyloid within selective pyramidal neurons, neurites and synapses in AD transgenic mice as plaques arise. CONCLUSION: The data support that Aß42 accumulation and aggregation begin within AD-vulnerable neurons in the brain. Progressive intraneuronal Aß42 aggregation disrupts the normal cytoarchitecture of neurites.


Assuntos
Doença de Alzheimer/patologia , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Placa Amiloide/patologia , Sinapses/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Humanos , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Placa Amiloide/metabolismo
16.
Front Aging Neurosci ; 14: 896848, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35783126

RESUMO

Evidence suggests that lightly myelinated cortical regions are vulnerable to aging and Alzheimer's disease (AD). However, it remains unknown whether plasma markers of amyloid and neurodegeneration are related to deficits in intracortical myelin content, and whether this relationship, in turn, is associated with altered patterns of resting-state functional connectivity (rs-FC). To shed light into these questions, plasma levels of amyloid-ß fragment 1-42 (Aß1-42) and neurofilament light chain (NfL) were measured using ultra-sensitive single-molecule array (Simoa) assays, and the intracortical myelin content was estimated with the ratio T1-weigthed/T2-weighted (T1w/T2w) in 133 cognitively normal older adults. We assessed: (i) whether plasma Aß1-42 and/or NfL levels were associated with intracortical myelin content at different cortical depths and (ii) whether cortical regions showing myelin reductions also exhibited altered rs-FC patterns. Surface-based multiple regression analyses revealed that lower plasma Aß1-42 and higher plasma NfL were associated with lower myelin content in temporo-parietal-occipital regions and the insular cortex, respectively. Whereas the association with Aß1-42 decreased with depth, the NfL-myelin relationship was most evident in the innermost layer. Older individuals with higher plasma NfL levels also exhibited altered rs-FC between the insula and medial orbitofrontal cortex. Together, these findings establish a link between plasma markers of amyloid/neurodegeneration and intracortical myelin content in cognitively normal older adults, and support the role of plasma NfL in boosting aberrant FC patterns of the insular cortex, a central brain hub highly vulnerable to aging and neurodegeneration.

17.
Front Aging Neurosci ; 14: 844534, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35493929

RESUMO

Glial cells are essential to understand Alzheimer's disease (AD) progression, given their role in neuroinflammation and neurodegeneration. There is a need for reliable and easy to manipulate models that allow studying the mechanisms behind neuron and glia communication. Currently available models such as co-cultures require complex methodologies and/or might not be affordable for all laboratories. With this in mind, we aimed to establish a straightforward in vitro setting with neurons and glial cells to study AD. We generated and optimized a 2D triple co-culture model with murine astrocytes, neurons and microglia, based on sequential seeding of each cell type. Immunofluorescence, western blot and ELISA techniques were used to characterize the effects of oligomeric Aß (oAß) in this model. We found that, in the triple co-culture, microglia increased the expression of anti-inflammatory marker Arginase I, and reduced pro-inflammatory iNOS and IL-1ß, compared with microglia alone. Astrocytes reduced expression of pro-inflammatory A1 markers AMIGO2 and C3, and displayed a ramified morphology resembling physiological conditions. Anti-inflammatory marker TGF-ß1 was also increased in the triple co-culture. Lastly, neurons increased post-synaptic markers, and developed more and longer branches than in individual primary cultures. Addition of oAß in the triple co-culture reduced synaptic markers and increased CD11b in microglia, which are hallmarks of AD. Consequently, we developed a straightforward and reproducible triple co-cultured model, where cells resemble physiological conditions better than in individual primary cultures: microglia are less inflammatory, astrocytes are less reactive and neurons display a more mature morphology. Moreover, we are able to recapitulate Aß-induced synaptic loss and CD11b increase. This model emerges as a powerful tool to study neurodegeneration and neuroinflammation in the context of AD and other neurodegenerative diseases.

18.
Alzheimers Res Ther ; 14(1): 80, 2022 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-35676734

RESUMO

BACKGROUND: Validation of new biomarkers of Alzheimer disease (AD) is crucial for the successful development and implementation of treatment strategies. Additional to traditional AT(N) biomarkers, neuroinflammation biomarkers, such as translocator protein (TSPO) and cystine/glutamine antiporter system (xc-), could be considered when assessing AD progression. Herein, we report the longitudinal investigation of [18F]DPA-714 and [18F]FSPG for their ability to detect TSPO and xc- biomarkers, respectively, in the 5xFAD mouse model for AD. METHODS: Expression of TSPO and xc- system was assessed longitudinally (2-12 months of age) on 5xFAD mice and their respective controls by positron emission tomography (PET) imaging using radioligands [18F]DPA-714 and [18F]FSPG. In parallel, in the same mice, amyloid-ß plaque deposition was assessed with the amyloid PET radiotracer [18F]florbetaben. In vivo findings were correlated to ex vivo immunofluorescence staining of TSPO and xc- in microglia/macrophages and astrocytes on brain slices. Physiological changes of the brain tissue were assessed by magnetic resonance imaging (MRI) in 12-month-old mice. RESULTS: PET studies showed a significant increase in the uptake of [18F]DPA-714 and [18F]FSPG in the cortex, hippocampus, and thalamus in 5xFAD but not in WT mice over time. The results correlate with Aß plaque deposition. Ex vivo staining confirmed higher TSPO overexpression in both, microglia/macrophages and astrocytes, and overexpression of xc- in non-glial cells of 5xFAD mice. Additionally, the results show that Aß plaques were surrounded by microglia/macrophages overexpressing TSPO. MRI studies showed significant tissue shrinkage and microstructural alterations in 5xFAD mice compared to controls. CONCLUSIONS: TSPO and xc- overexpression can be assessed by [18F]DPA-714 and [18F]FSPG, respectively, and correlate with the level of Aß plaque deposition obtained with a PET amyloid tracer. These results position the two tracers as promising imaging tools for the evaluation of disease progression. Longitudinal in vivo study in the 5xFAD mouse model shows that TSPO and oxidative stress assessment through [18F]DPA-714 and [18F]FSPG-PET imaging, respectively, could serve as a potential tool for the evaluation of Alzheimer disease progression.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Doenças Neuroinflamatórias , Estresse Oxidativo , Tomografia por Emissão de Pósitrons/métodos , Receptores de GABA/metabolismo
19.
Cell Death Dis ; 13(3): 253, 2022 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-35306512

RESUMO

Amyloid beta (Aß)-mediated synapse dysfunction is an early event in Alzheimer's disease (AD) pathogenesis and previous studies suggest that NMDA receptor (NMDAR) dysregulation may contribute to these pathological effects. Although Aß peptides impair NMDAR expression and activity, the mechanisms mediating these alterations in the early stages of AD are unclear. Here, we observed that NMDAR subunit NR2B and PSD-95 levels were aberrantly upregulated and correlated with Aß42 load in human postsynaptic fractions of the prefrontal cortex in early stages of AD patients, as well as in the hippocampus of 3xTg-AD mice. Importantly, NR2B and PSD95 dysregulation was revealed by an increased expression of both proteins in Aß-injected mouse hippocampi. In cultured neurons, Aß oligomers increased the NR2B-containing NMDAR density in neuronal membranes and the NMDA-induced intracellular Ca2+ increase, in addition to colocalization in dendrites of NR2B subunit and PSD95. Mechanistically, Aß oligomers required integrin ß1 to promote synaptic location and function of NR2B-containing NMDARs and PSD95 by phosphorylation through classic PKCs. These results provide evidence that Aß oligomers modify the contribution of NR2B to NMDAR composition and function in the early stages of AD through an integrin ß1 and PKC-dependent pathway. These data reveal a novel role of Aß oligomers in synaptic dysfunction that may be relevant to early-stage AD pathogenesis.


Assuntos
Doença de Alzheimer , Proteína Quinase C/metabolismo , Receptores de N-Metil-D-Aspartato , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Hipocampo/metabolismo , Humanos , Integrina beta1/metabolismo , Camundongos , N-Metilaspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo
20.
Parkinsonism Relat Disord ; 96: 29-35, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35149357

RESUMO

INTRODUCTION: Blood and cerebrospinal fluid represent emerging candidate fluids for biomarker identification in Parkinson's disease (PD). METHODS: We studied 8 individuals carrying the E46K-SNCA mutation (3 PD dementia (PDD), 1 tremor-dominant PD, 2 young rigid-akinetic PD and 2 asymptomatic) and 8 age- and sex-matched healthy controls. We quantified the levels of total alpha-synuclein (a-syn), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), Tau and ubiquitin carboxy-terminal hydrolase L1 (UCHL1) with SiMoA (Quanterix) in cerebrospinal fluid (CSF) of mutation carriers and in serum of all participants. The correlation between the concentration of biofluid markers and clinical outcomes was evaluated. RESULTS: Although based on a small number of cases, CSF a-syn was decreased in symptomatic E46K-SNCA carriers compared to the asymptomatic ones. Asymptomatic carriers exhibited similar serum biomarker levels as compared to matched controls, except for serum a-syn, which was higher in asymptomatic individuals. Carriers with PDD diagnosis displayed increased levels of serum NfL and GFAP compared to matched controls. These findings highly correlated with cognitive and motor status of E46K-SNCA carriers, but not with disease duration. CONCLUSIONS: Patients with familial forms of neurodegenerative disease exhibit variable penetrance of the phenotype and are exceptionally valuable for delineating biomarkers. Serum and CSF molecular biomarkers in E46K-SNCA mutation carriers show that a-syn might be suitable to track the conversion from asymptomatic to PD, whereas NfL and GFAP might serve to foresee the progression to PD dementia. These findings should be interpreted with caution and need to be replicated in other genetic synucleinopathy cohorts.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Parkinson , alfa-Sinucleína , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Humanos , Mutação , Doenças Neurodegenerativas/sangue , Doenças Neurodegenerativas/líquido cefalorraquidiano , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/genética , Doença de Parkinson/sangue , Doença de Parkinson/líquido cefalorraquidiano , Doença de Parkinson/diagnóstico , Doença de Parkinson/genética , alfa-Sinucleína/sangue , alfa-Sinucleína/líquido cefalorraquidiano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA