Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurosci ; 43(26): 4926-4940, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37236808

RESUMO

The key pathologic entities driving the destruction of synaptic function and integrity during the evolution of Alzheimer's disease (AD) remain elusive. Astrocytes are structurally and functionally integrated within synaptic and vascular circuitry and use calcium-based physiology to modulate basal synaptic transmission, vascular dynamics, and neurovascular coupling, which are central to AD pathogenesis. We used high-resolution multiphoton imaging to quantify all endogenous calcium signaling arising spontaneously throughout astrocytic somata, primary processes, fine processes, and capillary endfeet in the brain of awake APP/PS1 transgenic mice (11 male and 6 female mice). Endogenous calcium signaling within capillary endfeet, while surprisingly as active as astrocytic fine processes, was reduced ∼50% in the brain of awake APP/PS1 mice. Cortical astrocytes, in the presence of amyloid plaques in awake APP/PS1 mice, had a cell-wide increase in intracellular calcium associated with an increased frequency, amplitude, and duration of spontaneous calcium signaling. The cell-wide astrocytic calcium dysregulation was not directly related to distance to amyloid plaques. We could re-create the cell-wide intracellular calcium dysregulation in the absence of amyloid plaques following acute exposure to neuronally derived soluble Abeta from Tg2576 transgenic mice, in the living brain of male C57/Bl6 mice. Our findings highlight a role for astrocytic calcium pathophysiology in soluble-Abeta mediated neurodegenerative processes in AD. Additionally, therapeutic strategies aiming to protect astrocytic calcium physiology from soluble Abeta-mediated toxicity may need to pharmacologically enhance calcium signaling within the hypoactive capillary endfeet while reducing the hyperactivity of spontaneous calcium signaling throughout the rest of the astrocyte.SIGNIFICANCE STATEMENT Astrocytic calcium signaling is functionally involved in central pathologic processes of Alzheimer's disease. We quantified endogenous calcium signaling arising spontaneously in the brain of awake APP/PS1 mice, as general anesthesia suppressed astrocytic calcium signaling. Cell-wide astrocytic calcium dysregulation was not related to distance to amyloid plaques but mediated in part by neuronally derived soluble Abeta, supporting a role for astrocytes in soluble-Abeta mediated neurodegeneration. Spontaneous calcium signaling is largely compartmentalized and capillary endfeet were as active as fine processes but hypoactive in the presence of amyloid plaques, while the rest of the astrocyte became hyperactive. The cell-wide calcium pathophysiology in astrocytes may require a combination therapeutic strategy for hypoactive endfeet and astrocytic hyperactivity.


Assuntos
Doença de Alzheimer , Camundongos , Masculino , Feminino , Animais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Cálcio , Astrócitos/fisiologia , Placa Amiloide/patologia , Camundongos Transgênicos , Precursor de Proteína beta-Amiloide/genética , Modelos Animais de Doenças
2.
Proc Natl Acad Sci U S A ; 112(51): 15556-61, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26644572

RESUMO

Although the clustering of GFAP immunopositive astrocytes around amyloid-ß plaques in Alzheimer's disease has led to the widespread assumption that plaques attract astrocytes, recent studies suggest that astrocytes stay put in injury. Here we reexamine astrocyte migration to plaques, using quantitative spatial analysis and computer modeling to investigate the topology of astrocytes in 3D images obtained by two-photon microscopy of living APP/PS1 mice and WT littermates. In WT mice, cortical astrocyte topology fits a model in which a liquid of hard spheres exclude each other in a confined space. Plaques do not disturb this arrangement except at very large plaque loads, but, locally, cause subtle outward shifts of the astrocytes located in three tiers around plaques. These data suggest that astrocytes respond to plaque-induced neuropil injury primarily by changing phenotype, and hence function, rather than location.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/fisiologia , Placa Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/patologia , Fenômenos Biofísicos , Movimento Celular , Simulação por Computador , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Modelos Neurológicos , Placa Amiloide/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
Ann Neurol ; 78(2): 193-210, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26032020

RESUMO

OBJECTIVE: Migraine is among the most common and debilitating neurological conditions. Familial hemiplegic migraine type 1 (FHM1), a monogenic migraine subtype, is caused by gain-of-function of voltage-gated CaV 2.1 calcium channels. FHM1 mice carry human pathogenic mutations in the α1A subunit of CaV 2.1 channels and are highly susceptible to cortical spreading depression (CSD), the electrophysiologic event underlying migraine aura. To date, however, the mechanism underlying increased CSD/migraine susceptibility remains unclear. METHODS: We employed in vivo multiphoton microscopy of the genetically encoded Ca(2+)-indicator yellow cameleon to investigate synaptic morphology and [Ca(2+)]i in FHM1 mice. To study CSD-induced cerebral oligemia, we used in vivo laser speckle flowmetry and multimodal imaging. With electrophysiologic recordings, we investigated the effect of the CaV 2.1 gating modifier tert-butyl dihydroquinone on CSD in vivo. RESULTS: FHM1 mutations elevate neuronal [Ca(2+)]i and alter synaptic morphology as a mechanism for enhanced CSD susceptibility that we were able to normalize with a CaV 2.1 gating modifier in hyperexcitable FHM1 mice. At the synaptic level, axonal boutons were larger, and dendritic spines were predominantly of the mushroom type, which both provide a structural correlate for enhanced neuronal excitability. Resting neuronal [Ca(2+)]i was elevated in FHM1, with loss of compartmentalization between synapses and neuronal shafts. The percentage of calcium-overloaded neurons was increased. Neuronal [Ca(2+)]i surge during CSD was faster and larger, and post-CSD oligemia and hemoglobin desaturation were more severe in FHM1 brains. INTERPRETATION: Our findings provide a mechanism for enhanced CSD susceptibility in hemiplegic migraine. Abnormal synaptic Ca(2+) homeostasis and morphology may contribute to chronic neurodegenerative changes as well as enhanced vulnerability to ischemia in migraineurs.


Assuntos
Canais de Cálcio Tipo N/genética , Cálcio/metabolismo , Córtex Cerebral/metabolismo , Depressão Alastrante da Atividade Elétrica Cortical/genética , Enxaqueca com Aura/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Canais de Cálcio Tipo N/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Homeostase/genética , Hidroquinonas/farmacologia , Camundongos , Microscopia de Fluorescência por Excitação Multifotônica , Enxaqueca com Aura/genética , Enxaqueca com Aura/patologia , Mutação , Neurônios/efeitos dos fármacos , Neurônios/patologia , Sinapses/efeitos dos fármacos , Sinapses/patologia
4.
Cell Mol Neurobiol ; 36(2): 181-94, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26993512

RESUMO

The lymphatic clearance pathways of the brain are different compared to the other organs of the body and have been the subject of heated debates. Drainage of brain extracellular fluids, particularly interstitial fluid (ISF) and cerebrospinal fluid (CSF), is not only important for volume regulation, but also for removal of waste products such as amyloid beta (Aß). CSF plays a special role in clinical medicine, as it is available for analysis of biomarkers for Alzheimer's disease. Despite the lack of a complete anatomical and physiological picture of the communications between the subarachnoid space (SAS) and the brain parenchyma, it is often assumed that Aß is cleared from the cerebral ISF into the CSF. Recent work suggests that clearance of the brain mainly occurs during sleep, with a specific role for peri- and para-vascular spaces as drainage pathways from the brain parenchyma. However, the direction of flow, the anatomical structures involved and the driving forces remain elusive, with partially conflicting data in literature. The presence of Aß in the glia limitans in Alzheimer's disease suggests a direct communication of ISF with CSF. Nonetheless, there is also the well-described pathology of cerebral amyloid angiopathy associated with the failure of perivascular drainage of Aß. Herein, we review the role of the vasculature and the impact of vascular pathology on the peri- and para-vascular clearance pathways of the brain. The different views on the possible routes for ISF drainage of the brain are discussed in the context of pathological significance.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/patologia , Circulação Cerebrovascular , Linfa/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia , Animais , Humanos , Modelos Biológicos , Doenças Neurodegenerativas/líquido cefalorraquidiano
5.
J Neurosci ; 32(9): 3176-92, 2012 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-22378890

RESUMO

Amyloid ß (Aß) peptides, the main pathological species associated with Alzheimer's disease (AD), disturb intracellular calcium homeostasis, which in turn activates the calcium-dependent phosphatase calcineurin (CaN). CaN activation induced by Aß leads to pathological morphological changes in neurons, and overexpression of constitutively active calcineurin is sufficient to generate a similar phenotype, even without Aß. Here, we tested the hypothesis that calcineurin mediates neurodegenerative effects via activation of the nuclear transcription factor of activated T-cells (NFAT). We found that both spine loss and dendritic branching simplification induced by Aß exposure were mimicked by constitutively active NFAT, and abolished when NFAT activation was blocked using the genetically encoded inhibitor VIVIT. When VIVIT was specifically addressed to the nucleus, identical beneficial effects were observed, thus enforcing the role of NFAT transcriptional activity in Aß-related neurotoxicity. In vivo, when VIVIT or its nuclear counterpart were overexpressed in a transgenic model of Alzheimer's disease via a gene therapy approach, the spine loss and neuritic abnormalities observed in the vicinity of amyloid plaques were blocked. Overall, these results suggest that NFAT/calcineurin transcriptional cascades contribute to Aß synaptotoxicity, and may provide a new specific set of pathways for neuroprotective strategies.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Modelos Animais de Doenças , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/fisiologia , Transdução de Sinais/fisiologia , Doença de Alzheimer/patologia , Animais , Dendritos/patologia , Dendritos/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Inibição Neural/fisiologia , Oligopeptídeos/genética , Oligopeptídeos/farmacologia
6.
Acta Neuropathol ; 126(3): 353-64, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23818064

RESUMO

The interstitial fluid (ISF) drainage pathway has been hypothesized to underlie the clearance of solutes and metabolites from the brain. Previous work has implicated the perivascular spaces along arteries as the likely route for ISF clearance; however, it has never been demonstrated directly. The accumulation of amyloid ß (Aß) peptides in brain parenchyma is one of the pathological hallmarks of Alzheimer disease (AD), and it is likely related to an imbalance between production and clearance of the peptide. Aß drainage along perivascular spaces has been postulated to be one of the mechanisms that mediate the peptide clearance from the brain. We therefore devised a novel method to visualize solute clearance in real time in the living mouse brain using laser guided bolus dye injections and multiphoton imaging. This methodology allows high spatial and temporal resolution and revealed the kinetics of ISF clearance. We found that the ISF drains along perivascular spaces of arteries and capillaries but not veins, and its clearance exhibits a bi-exponential profile. ISF drainage requires a functional vasculature, as solute clearance decreased when perfusion was impaired. In addition, reduced solute clearance was observed in transgenic mice with significant vascular amyloid deposition; we suggest the existence of a feed-forward mechanism, by which amyloid deposition promotes further amyloid deposition. This important finding provides a mechanistic link between cerebrovascular disease and Alzheimer disease and suggests that facilitation of Aß clearance along the perivascular pathway should be considered as a new target for therapeutic approaches to Alzheimer disease and cerebral amyloid angiopathy.


Assuntos
Doença de Alzheimer/terapia , Drenagem , Líquido Extracelular/metabolismo , Isquemia/terapia , Acidente Vascular Cerebral/terapia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Capilares/patologia , Angiopatia Amiloide Cerebral/metabolismo , Modelos Animais de Doenças , Isquemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Acidente Vascular Cerebral/fisiopatologia
7.
J Neurosci ; 30(7): 2636-49, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20164348

RESUMO

Amyloid beta (Abeta)-containing plaques are surrounded by dystrophic neurites in the Alzheimer's disease (AD) brain, but whether and how plaques induce these neuritic abnormalities remain unknown. We tested the hypothesis that soluble oligomeric assemblies of Abeta, which surround plaques, induce calcium-mediated secondary cascades that lead to dystrophic changes in local neurites. We show that soluble Abeta oligomers lead to activation of the calcium-dependent phosphatase calcineurin (CaN) (PP2B), which in turn activates the transcriptional factor nuclear factor of activated T cells (NFAT). Activation of these signaling pathways, even in the absence of Abeta, is sufficient to produce a virtual phenocopy of Abeta-induced dystrophic neurites, dendritic simplification, and dendritic spine loss in both neurons in culture and in the adult mouse brain. Importantly, the morphological deficits in the vicinity of Abeta deposits in a mouse model of AD are ameliorated by CaN inhibition, supporting the hypothesis that CaN-NFAT are aberrantly activated by Abeta and that CaN-NFAT activation is responsible for disruption of neuronal structure near plaques. In accord with this, we also detect increased levels of an active form of CaN and NFATc4 in the nuclear fraction from the cortex of patients with AD. Thus, Abeta appears to mediate the neurodegeneration of AD, at least in part, by activation of CaN and subsequent NFAT-mediated downstream cascades.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Calcineurina/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neurônios/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Calcineurina/genética , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Espinhas Dendríticas , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Fatores de Transcrição NFATC/metabolismo , Neuritos , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Mudanças Depois da Morte , Transporte Proteico/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo
8.
Neuron ; 105(3): 549-561.e5, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31810839

RESUMO

Paravascular drainage of solutes, including ß-amyloid (Aß), appears to be an important process in brain health and diseases such as Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). However, the major driving force for clearance remains largely unknown. Here we used in vivo two-photon microscopy in awake head-fixed mice to assess the role of spontaneous vasomotion in paravascular clearance. Vasomotion correlated with paravascular clearance of fluorescent dextran from the interstitial fluid. Increasing the amplitude of vasomotion by means of visually evoked vascular responses resulted in increased clearance rates in the visual cortex of awake mice. Evoked vascular reactivity was impaired in mice with CAA, which corresponded to slower clearance rates. Our findings suggest that low-frequency arteriolar oscillations drive drainage of solutes. Targeting naturally occurring vasomotion in patients with CAA or AD may be a promising early therapeutic option for prevention of Aß accumulation in the brain.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Músculo Liso Vascular/irrigação sanguínea , Músculo Liso Vascular/metabolismo , Vigília/fisiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Capilares/metabolismo , Líquido Extracelular/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estimulação Luminosa/métodos , Sistema Vasomotor/metabolismo , Córtex Visual/irrigação sanguínea , Córtex Visual/metabolismo
9.
Sci Rep ; 9(1): 8964, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221985

RESUMO

Neuronal activity patterns are disrupted in neurodegenerative disorders, including Alzheimer's disease (AD). One example is disruption of corticothalamic slow oscillations responsible for sleep-dependent memory consolidation. Slow waves are periodic oscillations in neuronal activity occurring at frequencies of <1 Hz. The power, but not the frequency of slow oscillations is altered in a mouse model of AD. Optogenetic rescue of slow oscillations by increasing activity in cortical pyramidal neurons at the frequency of slow waves restores slow wave power, halts deposition of amyloid plaques and prevents neuronal calcium dysregulation. Here we determined whether driving this circuit at an increased rate would exacerbate the amyloid-dependent calcium dyshomeostasis in transgenic mice. Doubling the frequency of slow waves for one month with optogenetics resulted in increased amyloid beta - dependent disruptions in neuronal calcium homeostasis and loss of synaptic spines. Therefore, while restoration of physiological circuit dynamics is sufficient to abrogate the progression of Alzheimer's disease pathology and should be considered an avenue for clinical treatment of AD patients with sleep disorders, pathophysiological stimulation of neuronal circuits leads to activity - dependent acceleration of amyloid production, aggregation and downstream neuronal dysfunction.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Suscetibilidade a Doenças , Doença de Alzheimer/metabolismo , Amiloide/genética , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Imagem Molecular , Neurônios/metabolismo , Neurônios/patologia , Neurotransmissores/metabolismo , Placa Amiloide/etiologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Transmissão Sináptica
10.
Sci Rep ; 8(1): 4634, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29545579

RESUMO

Soluble amyloid ß oligomers (AßOs) are widely recognized neurotoxins that trigger aberrant signaling in specific subsets of neurons, leading to accumulated neuronal damage and memory disorders in Alzheimer's disease (AD). One of the profound downstream consequences of AßO-triggered events is dysregulation of cytosolic calcium concentration ([Ca2+]i), which has been implicated in synaptic failure, cytoskeletal abnormalities, and eventually neuronal death. We have developed an in vitro/in vivo drug screening assay to evaluate putative AßO-blocking candidates by measuring AßO-induced real-time changes in [Ca2+]i. Our screening assay demonstrated that the anti-AßO monoclonal antibody ACU3B3 exhibits potent blocking capability against a broad size range of AßOs. We showed that picomolar concentrations of AßOs were capable of increasing [Ca2+]i in primary neuronal cultures, an effect prevented by ACU3B3. Topical application of 5 nM AßOs onto exposed cortical surfaces also elicited significant calcium elevations in vivo, which was completely abolished by pre-treatment of the brain with 1 ng/mL (6.67 pM) ACU3B3. Our results provide strong support for the utility of this functional screening assay in identifying and confirming the efficacy of AßO-blocking drug candidates such as the human homolog of ACU3B3, which may emerge as the first experimental AD therapeutic to validate the amyloid oligomer hypothesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Encéfalo/efeitos dos fármacos , Cálcio/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Idoso de 80 Anos ou mais , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Mol Neurodegener ; 12(1): 27, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28327181

RESUMO

BACKGROUND: Amyloid-ß oligomers (oAß) are thought to mediate neurotoxicity in Alzheimer's disease (AD), and previous studies in AD transgenic mice suggest that calcium dysregulation may contribute to these pathological effects. Even though AD mouse models remain a valuable resource to investigate amyloid neurotoxicity, the concomitant presence of soluble Aß species, fibrillar Aß, and fragments of amyloid precursor protein (APP) complicate the interpretation of the phenotypes. METHOD: To explore the specific contribution of soluble oligomeric Aß (oAß) to calcium dyshomeostasis and synaptic morphological changes, we acutely exposed the healthy mouse brain, at 3 to 6 months of age, to naturally occurring soluble oligomers and investigated their effect on calcium levels using in vivo multiphoton imaging. RESULTS: We observed a dramatic increase in the levels of neuronal resting calcium, which was dependent upon extracellular calcium influx and activation of NMDA receptors. Ryanodine receptors, previously implicated in AD models, did not appear to be primarily involved using this experimental setting. We used the high resolution cortical volumes acquired in-vivo to measure the effect on synaptic densities and observed that, while spine density remained stable within the first hour of oAß exposure, a significant decrease in the number of dendritic spines was observed 24 h post treatment, despite restoration of intraneuronal calcium levels at this time point. CONCLUSIONS: These observations demonstrate a specific effect of oAß on NMDA-mediated calcium influx, which triggers synaptic collapse in vivo. Moreover, this work leverages a method to quantitatively measure calcium concentration at the level of neuronal processes, cell bodies and single synaptic elements repeatedly and thus can be applicable to testing putative drugs and/or other intervention methodologies.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/toxicidade , Encéfalo/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Sinapses/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Modelos Animais de Doenças , Humanos , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Sinapses/efeitos dos fármacos
12.
Neurobiol Aging ; 36(11): 2963-2971, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26248866

RESUMO

Cerebral amyloid angiopathy (CAA), the deposition of amyloid-ß in cerebrovascular walls, is the most common cause of lobar hemorrhagic stroke. Previous studies show that cerebrovascular amyloid-ß induces expression and activation of matrix metalloproteinase 9 (MMP-9) in cerebral vessels of amyloid precursor protein transgenic mice. Here, we extended these findings and evaluated MMP-9 expression in postmortem brain tissues of human CAA cases. MMP-9 colocalized with CAA, correlated with the severity of the vascular pathology, and was detected in proximity to microbleeds. We characterized a novel assay using longitudinal multiphoton microscopy and a novel tracer to visualize and quantify the magnitude and kinetics of hemorrhages in three dimensions in living mouse brains. We demonstrated that topical application of recombinant MMP-9 resulted in a time- and dose-dependent cerebral hemorrhage. Amyloid precursor protein mice with significant CAA developed more extensive hemorrhages which also appeared sooner after exposure to MMP-9. Our data suggest an important role for MMP-9 in development of hemorrhages in the setting of CAA. Inhibition of MMP-9 may present a preventive strategy for CAA-associated hemorrhage.


Assuntos
Angiopatia Amiloide Cerebral/complicações , Angiopatia Amiloide Cerebral/genética , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/genética , Metaloproteinase 9 da Matriz/metabolismo , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Animais , Angiopatia Amiloide Cerebral/metabolismo , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/prevenção & controle , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Metaloproteinase 9 da Matriz/farmacologia , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência por Excitação Multifotônica , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Proteínas Recombinantes/farmacologia , Índice de Gravidade de Doença
13.
Sci Transl Med ; 5(212): 212ra161, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24259049

RESUMO

Inheritance of the ε4 allele of apolipoprotein E (APOE) is the strongest genetic risk factor associated with the sporadic form of Alzheimer's disease (AD), whereas the rare APOE ε2 allele has the opposite effect. However, the mechanisms whereby APOE confers risk and protection remain uncertain. We used a gene transfer approach to bathe the cortex of amyloid plaque-bearing transgenic mice with virally expressed human APOE. We monitored amyloid-ß (Aß) with multiphoton imaging, in vivo microdialysis, and postmortem array tomography to study the kinetics of human APOE-mediated changes in Aß-related neurotoxicity in a mouse model of AD. We observed that human APOE4 increased the concentrations of oligomeric Aß within the interstitial fluid and exacerbated plaque deposition; the converse occurred after exposure to human APOE2. Peri-plaque synapse loss and dystrophic neurites were also worsened by APOE4 or attenuated by APOE2. Egress of Aß from the central nervous system (CNS) into the plasma was diminished by APOE3 and APOE4 compared to APOE2, in accord with isoform-specific retention of Aß in the CNS. Overall, our data show a differential effect of human APOE isoforms on amyloid deposition and clearance in transgenic mice and, more importantly, on Aß-mediated synaptotoxicity. These results suggest that the APOE genetic risk is mediated by Aß, and that therapeutic approaches aimed at decreasing APOE4, or increasing APOE2, may be beneficial in AD.


Assuntos
Amiloide/metabolismo , Apolipoproteínas E/genética , Encéfalo/metabolismo , Transfecção , Amiloide/toxicidade , Animais , Apolipoproteínas E/administração & dosagem , Humanos , Injeções Intraventriculares , Camundongos , Camundongos Transgênicos
14.
J Neuropathol Exp Neurol ; 71(11): 1009-17, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23095848

RESUMO

Cerebral amyloid angiopathy (CAA), the accumulation of ß-amyloid (Aß) in the walls of leptomeningeal and cortical blood vessels of the brain, is a major cause of intracerebral hemorrhage and cognitive impairment and is commonly associated with Alzheimer disease. The progression of CAA, as measured in transgenic mice by longitudinal imaging with multiphoton microscopy, occurs in a predictable linear manner. The dynamics of Aß deposition in and clearance from vascular walls and their relationship to the concentration of Aß in the brain are poorly understood. We manipulated Aß levels in the brain using 2 approaches: peripheral clearance via administration of the amyloid binding "peripheral sink" protein gelsolin and direct inhibition of its formation via administration of LY-411575, a small-molecule γ-secretase inhibitor. We found that gelsolin and LY-411575 both reduced the rate of CAA progression in Tg2576 mice from untreated rates of 0.58% ± 0.15% and 0.52% ± 0.09% to 0.11% ± 0.18% (p = 0.04) and -0.17% ± 0.09% (p < 0.001) of affected vessel per day, respectively, in the absence of an immune response. The progression of CAA was also halted when gelsolin was combined with LY-411575 (-0.004% ± 0.10%, p < 0.003). These data suggest that CAA progression can be prevented with non-immune approaches that may reduce the availability of soluble Aß but without evidence of substantial amyloid clearance from vessels.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/fisiologia , Angiopatia Amiloide Cerebral/prevenção & controle , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/prevenção & controle , Animais , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/patologia , Progressão da Doença , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Solubilidade
15.
J Alzheimers Dis ; 22(2): 469-82, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20847413

RESUMO

Among the different paradigms aimed at interfering with amyloid-ß (Aß)-related pathology, the attenuation of amyloid-ß protein precursor (AßPP) processing to limit Aß levels seems to be a promising one. Along with the development of BACE1 inhibitors, and the generation of its knock-out mice, accumulating data raise concerns regarding a total inhibition of the enzyme as it shares the processing of other substrates. We described a novel approach to interfere with the specific interaction between AßPP and BACE1 using monoclonal antibodies directed to the ß-secretase cleavage site upon the substrate, AßPP. Such antibodies limit AßPP cleavage in a cellular model of Alzheimer's disease (AD) and avoid the total inhibition of BACE1. Here, we demonstrate the ability of AßPP ß-site antibodies to interfere with Aß production in vivo. Systemic antibody treatment diminished Aß plaques, membrane-associated oligomers, and intracellular Aß accumulation, all of which have been implicated in cellular death and synaptic loss, suggesting that this approach may be an applicable strategy for AD treatment.


Assuntos
Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Anticorpos Monoclonais/uso terapêutico , Encéfalo/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/imunologia , Animais , Ácido Aspártico Endopeptidases/metabolismo , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa