Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 10868, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740836

RESUMEN

Therapeutic antibodies have been developed to target amyloid-beta (Aß), and some of these slow the progression of Alzheimer's disease (AD). However, they can also cause adverse events known as amyloid-related imaging abnormalities with edema (ARIA-E). We investigated therapeutic Aß antibody binding to cerebral amyloid angiopathy (CAA) fibrils isolated from human leptomeningeal tissue to study whether this related to the ARIA-E frequencies previously reported by clinical trials. The binding of Aß antibodies to CAA Aß fibrils was evaluated in vitro using immunoprecipitation, surface plasmon resonance, and direct binding assay. Marked differences in Aß antibody binding to CAA fibrils were observed. Solanezumab and crenezumab showed negligible CAA fibril binding and these antibodies have no reported ARIA-E cases. Lecanemab showed a low binding to CAA fibrils, consistent with its relatively low ARIA-E frequency of 12.6%, while aducanumab, bapineuzumab, and gantenerumab all showed higher binding to CAA fibrils and substantially higher ARIA-E frequencies (25-35%). An ARIA-E frequency of 24% was reported for donanemab, and its binding to CAA fibrils correlated with the amount of pyroglutamate-modified Aß present. The findings of this study support the proposal that Aß antibody-CAA interactions may relate to the ARIA-E frequency observed in patients treated with Aß-based immunotherapies.


Asunto(s)
Péptidos beta-Amiloides , Angiopatía Amiloide Cerebral , Humanos , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Unión Proteica , Amiloide/metabolismo , Amiloide/inmunología , Resonancia por Plasmón de Superficie
2.
J Neuroinflammation ; 17(1): 366, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261619

RESUMEN

BACKGROUND: The ability to distinguish resident microglia from infiltrating myeloid cells by flow cytometry-based surface phenotyping is an important technique for examining age-related neuroinflammation. The most commonly used surface markers for the identification of microglia include CD45 (low-intermediate expression), CD11b, Tmem119, and P2RY12. METHODS: In this study, we examined changes in expression levels of these putative microglia markers in in vivo animal models of stroke, cerebral amyloid angiopathy (CAA), and aging as well as in an ex vivo LPS-induced inflammation model. RESULTS: We demonstrate that Tmem119 and P2RY12 expression is evident within both CD45int and CD45high myeloid populations in models of stroke, CAA, and aging. Interestingly, LPS stimulation of FACS-sorted adult microglia suggested that these brain-resident myeloid cells can upregulate CD45 and downregulate Tmem119 and P2RY12, making them indistinguishable from peripherally derived myeloid populations. Importantly, our findings show that these changes in the molecular signatures of microglia can occur without a contribution from the other brain-resident or peripherally sourced immune cells. CONCLUSION: We recommend future studies approach microglia identification by flow cytometry with caution, particularly in the absence of the use of a combination of markers validated for the specific neuroinflammation model of interest. The subpopulation of resident microglia residing within the "infiltrating myeloid" population, albeit small, may be functionally important in maintaining immune vigilance in the brain thus should not be overlooked in neuroimmunological studies.


Asunto(s)
Biomarcadores/análisis , Citometría de Flujo/métodos , Inflamación/inmunología , Inflamación/patología , Microglía , Envejecimiento/inmunología , Envejecimiento/patología , Animales , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología
3.
Neurobiol Dis ; 139: 104823, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32119976

RESUMEN

The DNA vaccine, AV-1959D, targeting N-terminal epitope of Aß peptide, has been proven immunogenic in mice, rabbits, and non-human primates, while its therapeutic efficacy has been shown in mouse models of Alzheimer's disease (AD). Here we report for the first time on IND-enabling biodistribution and safety/toxicology studies of cGMP-grade AV-1959D vaccine in the Tg2576 mouse model of AD. We also tested acute neuropathology safety profiles of AV-1959D in another AD disease model, Tg-SwDI mice with established vascular and parenchymal Aß pathology in a pre-clinical translational study. Biodistribution studies two days after the injection demonstrated high copy numbers of AV-1959D plasmid after single immunization of Tg2576 mice at the injection sites but not in the tissues of distant organs. Plasmids persisted at the injection sites of some mice 60 days after vaccination. In Tg2576 mice with established amyloid pathology, we did not observe short- or long-term toxicities after multiple immunizations with three doses of AV-1959D. Assessment of the repeated dose acute safety of AV-1959D in cerebral amyloid angiopathy (CAA) prone Tg-SwDI mice did not reveal any immunotherapy-induced vasogenic edema detected by magnetic resonance imaging (MRI) or increased microhemorrhages. Multiple immunizations of Tg-SwDI mice with AV-1959D did not induce T and B cell infiltration, glial activation, vascular deposition of Aß, or neuronal degeneration (necrosis and apoptosis) greater than that in the control group determined by immunohistochemistry of brain tissues. Taken together, the safety data from two different mouse models of AD substantiate a favorable safety profile of the cGMP grade AV-1959D vaccine supporting its progression to first-in-human clinical trials.


Asunto(s)
Vacunas contra el Alzheimer/inmunología , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Formación de Anticuerpos , Angiopatía Amiloide Cerebral/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo
4.
Acta Neuropathol Commun ; 7(1): 141, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477180

RESUMEN

Neuropathological follow-up of patients with Alzheimer's disease (AD) who participated in the first clinical trial of Amyloid-ß 42 (Aß42) immunization (AN1792, Elan Pharmaceuticals) has shown that immunization can induce removal of Aß42 and Aß40 from plaques, whereas analysis of the cerebral vessels has shown increased levels of these Aß peptides in cerebral amyloid angiopathy (CAA). Aß43 has been less frequently studied in AD, but its aggregation propensity and neurotoxic properties suggest it may have an important pathogenic role. In the current study we show by using immunohistochemistry that in unimmunized AD patients Aß43 is a frequent constituent of plaques (6.0% immunostained area), similar to Aß42 (3.9% immunostained area). Aß43 immunostained area was significantly higher than that of Aß40 (2.3%, p = 0.006). In addition, we show that Aß43 is only a minor component of CAA in both parenchymal vessels (1.5 Aß43-positive vessels per cm2 cortex vs. 5.3 Aß42-positive vessels, p = 0.03, and 6.2 Aß40-positive vessels, p = 0.045) and leptomeningeal vessels (5.6% Aß43-positive vessels vs. 17.3% Aß42-positive vessels, p = 0.007, and 27.4% Aß40-positive vessels, p = 0.003). Furthermore, we have shown that Aß43 is cleared from plaques after Aß immunotherapy, similar to Aß42 and Aß40. Cerebrovascular Aß43 levels did not change after immunotherapy.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Inmunización , Fragmentos de Péptidos/inmunología , Placa Amiloide/inmunología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Femenino , Humanos , Masculino , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo
5.
Front Immunol ; 10: 1534, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333665

RESUMEN

Cerebral amyloid angiopathy (CAA) corresponds to the deposition of amyloid material in the cerebral vasculature, leading to structural modifications of blood vessel walls. The most frequent form of sporadic CAA involves fibrillar ß-amyloid peptide (Aß) deposits, mainly the 40 amino acid form (Aß1-40), which are commonly found in the elderly with or without Alzheimer's disease. Sporadic CAA usually remains clinically silent. However, in some cases, acute complications either hemorrhagic or inflammatory can occur. Similar complications occurred after active or passive immunization against Aß in experimental animal models exhibiting CAA, and in subjects with Alzheimer's disease during clinical trials. The triggering of these adverse events by active immunization and monoclonal antibody administration in CAA-bearing individuals suggests that analogous mechanisms could be involved during spontaneous CAA complications, drawing particular attention to the role of anti-Aß antibodies. However, antibodies that react with several monomeric and aggregated forms of Aß spontaneously occur in virtually all human individuals, hence being part of the "natural antibody" repertoire. Natural antibodies are usually described as having low-affinity and high cross-reactivity toward microbial components and autoantigens. Although frequently of the IgM class, they also belong to IgG and IgA isotypes. They likely display homeostatic functions and protective roles in aging. Until recently, the peculiar properties of these natural antibodies have hindered proper analysis of the Aß-reactive antibody repertoire and the study of their implication in CAA complications. Herein, we review and comment the evidences of an auto-immune nature of spontaneous CAA complications, and discuss implications for forthcoming research and clinical practice.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales , Angiopatía Amiloide Cerebral , Inmunización Pasiva , Fragmentos de Péptidos/inmunología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Humanos
6.
J Neurosci ; 37(4): 936-959, 2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28123027

RESUMEN

Alzheimer's disease (AD) is characterized by the presence of parenchymal amyloid-ß (Aß) plaques, cerebral amyloid angiopathy (CAA) and neurofibrillary tangles. Currently there are no effective treatments for AD. Immunotherapeutic approaches under development are hampered by complications related to ineffectual clearance of CAA. Genome-wide association studies have demonstrated the importance of microglia in AD pathogenesis. Microglia are the primary innate immune cells of the brain. Depending on their activation state and environment, microglia can be beneficial or detrimental. In our prior work, we showed that stimulation of innate immunity with Toll-like receptor 9 agonist, class B CpG (cytosine-phosphate-guanine) oligodeoxynucleotides (ODNs), can reduce amyloid and tau pathologies without causing toxicity in Tg2576 and 3xTg-AD mouse models. However, these transgenic mice have relatively little CAA. In the current study, we evaluated the therapeutic profile of CpG ODN in a triple transgenic mouse model, Tg-SwDI, with abundant vascular amyloid, in association with low levels of parenchymal amyloid deposits. Peripheral administration of CpG ODN, both before and after the development of CAA, negated short-term memory deficits, as assessed by object-recognition tests, and was effective at improving spatial and working memory evaluated using a radial arm maze. These findings were associated with significant reductions of CAA pathology lacking adverse effects. Together, our extensive evidence suggests that this innovative immunomodulation may be a safe approach to ameliorate all hallmarks of AD pathology, supporting the potential clinical applicability of CpG ODN. SIGNIFICANCE STATEMENT: Recent genetic studies have underscored the emerging role of microglia in Alzheimer's disease (AD) pathogenesis. Microglia lose their amyloid-ß-clearing capabilities with age and as AD progresses. Therefore, the ability to modulate microglia profiles offers a promising therapeutic avenue for reducing AD pathology. Current immunotherapeutic approaches have been limited by poor clearance of a core AD lesion, cerebral amyloid angiopathy (CAA). The present study used Tg-SwDI mice, which have extensive CAA. We found that stimulation of the innate immune system and microglia/macrophage activation via Toll-like receptor 9 using CpG (cytosine-phosphate-guanine) oligodeoxynucleotides (ODNs) leads to cognitive improvements and CAA reduction, without associated toxicity. Our data indicate that this novel concept of immunomodulation represents a safer method to reduce all aspects of AD pathology and provide essential information for potential clinical use of CpG ODN.


Asunto(s)
Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/metabolismo , Cognición/fisiología , Inmunidad Innata/fisiología , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Cognición/efectos de los fármacos , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodesoxirribonucleótidos/farmacología , Oligodesoxirribonucleótidos/uso terapéutico , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/inmunología , Placa Amiloide/metabolismo , Receptor Toll-Like 9/agonistas
7.
J Neurol ; 264(2): 373-381, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28000005

RESUMEN

Brain magnetic resonance imaging (MRI) of patients with Alzheimer's disease (AD) sometimes reveals multiple cerebral microbleeds (CMBs) and confluent white matter hyperintensities (WMHs) similar to those observed in cerebral amyloid angiopathy-related inflammation (CAA-I). To determine whether there might be common pathophysiological mechanisms underlying the MRI findings of multiple CMBs and confluent WMHs, we investigated the cerebrospinal fluid (CSF) profiles of 38 AD, five amnestic mild cognitive impairment (MCI), and six CAA-I patients. The AD and MCI patients were divided into groups of patients with (n = 10) or without (n = 33) multiple CMBs (n ≥ 2) on T2*-gradient echo sequences of brain MRI. We compared the CSF profiles of AD and MCI patients with or without multiple CMBs, and CAA-I patients. The brain MRIs of the patients with multiple CMBs revealed severe degrees of WMHs compared with the patients without multiple CMBs. The levels of CSF anti-amyloid ß autoantibody and interleukin 8, and CSF/serum albumin ratios and immunoglobulin G indexes, were significantly higher in CAA-I patients than the other groups. However, there were no significant differences in the CSF profiles of patients with or without multiple CMBs. Our study provides evidence for different pathophysiological mechanisms underlying these differential MRI findings in AD and CAA-I.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Encéfalo/diagnóstico por imagen , Angiopatía Amiloide Cerebral/líquido cefalorraquídeo , Hemorragia Cerebral/líquido cefalorraquídeo , Disfunción Cognitiva/líquido cefalorraquídeo , Anciano , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Aterosclerosis/líquido cefalorraquídeo , Aterosclerosis/complicaciones , Aterosclerosis/inmunología , Autoanticuerpos/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Encéfalo/inmunología , Angiopatía Amiloide Cerebral/complicaciones , Angiopatía Amiloide Cerebral/diagnóstico por imagen , Angiopatía Amiloide Cerebral/inmunología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/inmunología , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/inmunología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Factores de Riesgo , Punción Espinal , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/inmunología
8.
Neuropathol Appl Neurobiol ; 43(6): 492-504, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27543695

RESUMEN

AIMS: Amyloid beta (Aß) accumulation in the walls of leptomeningeal arteries as cerebral amyloid angiopathy (CAA) is a major feature of Alzheimer's disease. In this study, we used global quantitative proteomic analysis to examine the hypothesis that the leptomeningeal arteries derived from patients with CAA have a distinct endophenotypic profile compared to those from young and elderly controls. METHODS: Freshly dissected leptomeningeal arteries from the Newcastle Brain Tissue Resource and Edinburgh Sudden Death Brain Bank from seven elderly (82.9 ± 7.5 years) females with severe capillary and arterial CAA, as well as seven elderly (88.3 ± 8.6 years) and five young (45.4 ± 3.9 years) females without CAA were used in this study. Arteries from four patients with CAA, two young and two elderly controls were individually analysed using quantitative proteomics. Key proteomic findings were then validated using immunohistochemistry. RESULTS: Bioinformatics interpretation of the results showed a significant enrichment of the immune response/classical complement and extracellular matrix remodelling pathways (P < 0.05) in arteries affected by CAA vs. those from young and elderly controls. Clusterin (apolipoprotein J) and tissue inhibitor of metalloproteinases-3 (TIMP3), validated using immunohistochemistry, were shown to co-localize with Aß and to be up-regulated in leptomeningeal arteries from CAA patients compared to young and elderly controls. CONCLUSIONS: Global proteomic profiling of brain leptomeningeal arteries revealed that clusterin and TIMP3 increase in leptomeningeal arteries affected by CAA. We propose that clusterin and TIMP3 could facilitate perivascular clearance and may serve as novel candidate therapeutic targets for CAA.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Angiopatía Amiloide Cerebral/metabolismo , Clusterina/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Arterias/metabolismo , Encéfalo/inmunología , Encéfalo/patología , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Vía Clásica del Complemento , Biología Computacional , Endofenotipos , Matriz Extracelular/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Proteómica
9.
Brain Behav Immun ; 54: 211-225, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26872418

RESUMEN

Cerebral beta-amyloidosis, one of the pathological hallmarks of Alzheimer's disease (AD), elicits a well-characterised, microglia-mediated local innate immune response. In contrast, it is not clear whether cells of the adaptive immune system, in particular T-cells, react to cerebral amyloidosis in AD. Even though parenchymal T-cells have been described in post-mortem brains of AD patients, it is not known whether infiltrating T-cells are specifically recruited to the extracellular deposits of beta-amyloid, and whether they are locally activated into proliferating, effector cells upon interaction with antigen-presenting cells (APCs). To address these issues we have analysed by confocal microscopy and flow-cytometry the localisation and activation status of both T-cells and APCs in transgenic (tg) mice models of AD-like cerebral amyloidosis. Increased numbers of infiltrating T-cells were found in amyloid-burdened brain regions of tg mice, with concomitant up-regulation of endothelial adhesion molecules ICAM-1 and VCAM-1, compared to non-tg littermates. The infiltrating T-cells in tg brains did not co-localise with amyloid plaques, produced less interferon-gamma than those in controls and did not proliferate locally. Bona-fide dendritic cells were virtually absent from the brain parenchyma of both non-tg and tg mice, and APCs from tg brains showed an immature phenotype, with accumulation of MHC-II in intracellular compartments. These results indicate that cerebral amyloidosis promotes T-cell infiltration but interferes with local antigen presentation and T-cell activation. The inability of the brain immune surveillance to orchestrate a protective immune response to amyloid-beta peptide might contribute to the accumulation of amyloid in the progression of the disease.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Angiopatía Amiloide Cerebral/inmunología , Linfocitos T/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/patología , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Encéfalo/metabolismo , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fenotipo , Placa Amiloide , Linfocitos T/metabolismo , Linfocitos T/patología , Regulación hacia Arriba
10.
Brain ; 139(Pt 2): 563-77, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26493635

RESUMEN

Prominent cerebral amyloid angiopathy is often observed in the brains of elderly individuals and is almost universally found in patients with Alzheimer's disease. Cerebral amyloid angiopathy is characterized by accumulation of the shorter amyloid-ß isoform(s) (predominantly amyloid-ß40) in the walls of leptomeningeal and cortical arterioles and is likely a contributory factor to vascular dysfunction leading to stroke and dementia in the elderly. We used transgenic mice with prominent cerebral amyloid angiopathy to investigate the ability of ponezumab, an anti-amyloid-ß40 selective antibody, to attenuate amyloid-ß accrual in cerebral vessels and to acutely restore vascular reactivity. Chronic administration of ponezumab to transgenic mice led to a significant reduction in amyloid and amyloid-ß accumulation both in leptomeningeal and brain vessels when measured by intravital multiphoton imaging and immunohistochemistry. By enriching for cerebral vascular elements, we also measured a significant reduction in the levels of soluble amyloid-ß biochemically. We hypothesized that the reduction in vascular amyloid-ß40 after ponezumab administration may reflect the ability of ponezumab to mobilize an interstitial fluid pool of amyloid-ß40 in brain. Acutely, ponezumab triggered a significant and transient increase in interstitial fluid amyloid-ß40 levels in old plaque-bearing transgenic mice but not in young animals. We also measured a beneficial effect on vascular reactivity following acute administration of ponezumab, even in vessels where there was a severe cerebral amyloid angiopathy burden. Taken together, the beneficial effects ponezumab administration has on reducing the rate of cerebral amyloid angiopathy deposition and restoring cerebral vascular health favours a mechanism that involves rapid removal and/or neutralization of amyloid-ß species that may otherwise be detrimental to normal vessel function.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/administración & dosificación , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Angiopatía Amiloide Cerebral/metabolismo , Endotelio Vascular/metabolismo , Inmunización Pasiva/métodos , Péptidos beta-Amiloides/inmunología , Animales , Angiopatía Amiloide Cerebral/inmunología , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Femenino , Masculino , Ratones , Ratones Transgénicos , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/inmunología , Placa Amiloide/metabolismo
11.
Curr Neurol Neurosci Rep ; 15(8): 54, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26096511

RESUMEN

Cerebral amyloid angiopathy-related inflammation (CAARI) is a recently recognized syndrome of reversible encephalopathy seen in a subset of patients with cerebral amyloid angiopathy (CAA). CAA is a disorder of the elderly in which amyloid peptides are deposited in the walls of cerebral arteries, leading to microhemorrhages, macrohemorrhages, and eventually dementia. In a few cases, the amyloid deposition is accompanied by inflammation or edema. The clinical syndrome of CAARI is distinguished by subacute neurobehavioral symptoms, headaches, seizures, and stroke-like signs, contrasting the acute intracranial hemorrhage typically seen in CAA. Magnetic resonance imaging findings may be symmetric or asymmetric and involve patchy or confluent T2 hyperintense lesions in the cortex and subcortical white matter. Recent diagnostic criteria have been proposed which help distinguish CAARI from alternative diagnoses. Improvement has been reported in most cases with immunosuppression, although a few cases have had recurrent symptoms. Here, we review the clinical and radiologic features of CAARI and compare these with CAA.


Asunto(s)
Angiopatía Amiloide Cerebral/complicaciones , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Diagnóstico Diferencial , Cefalea/etiología , Humanos , Inflamación/etiología , Inflamación/inmunología , Inflamación/patología , Imagen por Resonancia Magnética/métodos , Convulsiones/etiología , Accidente Cerebrovascular/etiología
12.
J Neurosci ; 35(16): 6265-76, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904780

RESUMEN

Alzheimer's disease (AD) and familial Danish dementia (FDD) are degenerative neurological diseases characterized by amyloid pathology. Normal human sera contain IgG antibodies that specifically bind diverse preamyloid and amyloid proteins and have shown therapeutic potential in vitro and in vivo. We cloned one of these antibodies, 3H3, from memory B cells of a healthy individual using a hybridoma method. 3H3 is an affinity-matured IgG that binds a pan-amyloid epitope, recognizing both Aß and λ Ig light chain (LC) amyloids, which are associated with AD and primary amyloidosis, respectively. The pan-amyloid-binding properties of 3H3 were demonstrated using ELISA, immunohistochemical studies, and competition binding assays. Functional studies showed that 3H3 inhibits both Aß and LC amyloid formation in vitro and abrogates disruption of hippocampal synaptic plasticity by AD-patient-derived soluble Aß in vivo. A 3H3 single-chain variable fragment (scFv) retained the binding specificity of the 3H3 IgG and, when expressed in the brains of transgenic mice using an adeno-associated virus (AAV) vector, decreased parenchymal Aß amyloid deposition in TgCRND8 mice and ADan (Danish Amyloid) cerebral amyloid angiopathy in the mouse model of FDD. These data indicate that naturally occurring human IgGs can recognize a conformational, amyloid-specific epitope and have potent anti-amyloid activities, providing a rationale to test their potential as antibody therapeutics for diverse neurological and other amyloid diseases.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Amiloide/metabolismo , Anticuerpos Monoclonales/inmunología , Inmunoglobulina G/inmunología , Amiloide/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Encéfalo/metabolismo , Catarata/inmunología , Ataxia Cerebelosa/inmunología , Angiopatía Amiloide Cerebral/inmunología , Sordera/inmunología , Demencia/inmunología , Humanos , Inmunoglobulina G/farmacología , Masculino , Ratones , Ratones Transgénicos , Ratas
13.
Neurodegener Dis ; 13(1): 38-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24021982

RESUMEN

BACKGROUND: Primary angiitis of the central nervous system (PACNS) is a rare but serious condition. A fraction of patients suffering from PACNS concurrently exhibit pronounced cerebral amyloid angiopathy (CAA) which is characterized by deposits of amyloid-ß (Aß) in and around the walls of small and medium-sized arteries of the brain. PACNS with CAA has been identified as a distinct disease entity, termed Aß-related angiitis (ABRA). Evidence points to an immune reaction to vessel wall Aß as the trigger of vasculitis. OBJECTIVE: To investigate whether the inflammatory response to Aß has (1) any effect on the status of immune activation in the brain parenchyma and (2) leads to clearance of Aß from brain parenchyma. METHODS: We studied immune activation and Aß load by quantitative immunohistochemical analysis in brain parenchyma adjacent to affected vessels in 11 ABRA patients and 10 matched CAA controls. RESULTS: ABRA patients showed significantly increased immune activation and decreased Aß loads in the brain parenchyma adjacent to affected vessels. CONCLUSION: Our results are in line with the hypothesis of ABRA being the result of an excessive immune response to Aß and show that this can lead to enhanced clearance of Aß from the brain parenchyma by immune-mediated mechanisms.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Encéfalo/patología , Vasculitis del Sistema Nervioso Central/inmunología , Vasculitis del Sistema Nervioso Central/patología , Anciano , Péptidos beta-Amiloides/análisis , Estudios de Casos y Controles , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/patología , Femenino , Humanos , Inmunohistoquímica , Activación de Macrófagos , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Placa Amiloide/inmunología , Placa Amiloide/patología
14.
Ann Neurol ; 73(4): 449-58, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23625526

RESUMEN

OBJECTIVE: Cerebral amyloid angiopathy-related inflammation (CAA-ri) is characterized by vasogenic edema and multiple cortical/subcortical microbleeds, sharing several aspects with the recently defined amyloid-related imaging abnormalities (ARIA) reported in Alzheimer's disease (AD) passive immunization therapies. Herein, we investigated the role of anti-amyloid ß (Aß) autoantibodies in the acute and remission phases of CAA-ri. METHODS: We used a novel ultrasensitive technique on patients from a retrospective multicenter case-control study, and evaluated the anti-Aß autoantibody concentration in the cerebrospinal fluid (CSF) of 10 CAA-ri, 8 CAA, 14 multiple sclerosis, and 25 control subjects. Levels of soluble Aß40, Aß42, tau, P-181 tau, and APOE genotype were also investigated. RESULTS: During the acute phase of CAA-ri, anti-Aß autoantibodies were specifically increased and directly correlated with Aß mobilization, together with augmented tau and P-181 tau. Following clinical and radiological remission, autoantibodies progressively returned to control levels, and both soluble Aß and axonal degeneration markers decreased in parallel. INTERPRETATION: Our data support the hypothesis that the pathogenesis of CAA-ri may be mediated by a selective autoimmune reaction against cerebrovascular Aß, directly related to autoantibody concentration and soluble Aß. The CSF dosage of anti-Aß autoantibodies with the technique here described can thus be proposed as a valid alternative tool for the diagnosis of CAA-ri. Moreover, given the similarities between ARIA developing spontaneously and those observed during immunization trials, anti-Aß autoantibodies can be considered as novel potential biomarkers in future amyloid-modifying therapies for the treatment of AD and CAA.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Autoanticuerpos/líquido cefalorraquídeo , Angiopatía Amiloide Cerebral , Inflamación , Adulto , Anciano , Péptidos beta-Amiloides/inmunología , Apolipoproteínas E/genética , Encéfalo/patología , Estudios de Casos y Controles , Angiopatía Amiloide Cerebral/líquido cefalorraquídeo , Angiopatía Amiloide Cerebral/complicaciones , Angiopatía Amiloide Cerebral/inmunología , Femenino , Humanos , Inflamación/líquido cefalorraquídeo , Inflamación/etiología , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/líquido cefalorraquídeo , Fosforilación , Estudios Retrospectivos , Esteroides/uso terapéutico , Proteínas tau/líquido cefalorraquídeo
15.
Proc Natl Acad Sci U S A ; 110(8): 3089-94, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382216

RESUMEN

Deposition of amyloid-ß (Aß) in cerebral arteries, known as cerebral amyloid angiopathy (CAA), occurs both in the setting of Alzheimer's disease and independent of it, and can cause cerebrovascular insufficiency and cognitive deficits. The mechanisms leading to CAA have not been established, and no therapeutic targets have been identified. We investigated the role of CD36, an innate immunity receptor involved in Aß trafficking, in the neurovascular dysfunction, cognitive deficits, and amyloid accumulation that occurs in mice expressing the Swedish mutation of the amyloid precursor protein (Tg2576). We found that Tg2576 mice lacking CD36 have a selective reduction in Aß1-40 and CAA. This reduced vascular amyloid deposition was associated with preservation of the Aß vascular clearance receptor LRP-1, and protection from the deleterious effects of Aß on cerebral arterioles. These beneficial vascular effects were reflected by marked improvements in neurovascular regulation and cognitive performance. Our data suggest that CD36 promotes vascular amyloid deposition and the resulting cerebrovascular damage, leading to neurovascular dysfunction and cognitive deficits. These findings identify a previously unrecognized role of CD36 in the mechanisms of vascular amyloid deposition, and suggest that this scavenger receptor is a putative therapeutic target for CAA and related conditions.


Asunto(s)
Antígenos CD36/inmunología , Angiopatía Amiloide Cerebral/inmunología , Inmunidad Innata , Animales , Vasos Sanguíneos/metabolismo , Antígenos CD36/genética , Circulación Cerebrovascular , Técnica del Anticuerpo Fluorescente , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Pericitos/inmunología , Proteína de la Zonula Occludens-1/metabolismo
16.
Aging Cell ; 12(3): 358-69, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23425004

RESUMEN

Several studies have shown that the accumulation of ß-amyloid peptides in the brain parenchyma or vessel wall generates an inflammatory environment. Some even suggest that there is a cause-and-effect relationship between inflammation and the development of Alzheimer's disease and/or cerebral amyloid angiopathy (CAA). Here, we studied the ability of wild-type Aß1-40 -peptide (the main amyloid peptide that accumulates in the vessel wall in sporadic forms of CAA) to modulate the phenotypic transition of vascular smooth muscle cells (VSMCs) toward an inflammatory/de-differentiated state. We found that Aß1-40 -peptide alone neither induces an inflammatory response, nor decreases the expression of contractile markers; however, the inflammatory response of VSMCs exposed to Aß1-40 -peptide prior to the addition of the pro-inflammatory cytokine IL-1ß is greatly intensified compared with IL-1ß-treated VSMCs previously un-exposed to Aß1-40 -peptide. Similar conclusions could be drawn when tracking the decline of contractile markers. Furthermore, we found that the mechanism of this potentiation highly depends on an Aß1-40 preactivation of the PI3 Kinase and possibly NFκB pathway; indeed, blocking the activation of these pathways during Aß1-40 -peptide treatment completely suppressed the observed potentiation. Finally, strengthening the possible in vivo relevance of our findings, we evidenced that endothelial cells exposed to Aß1-40 -peptide generate an inflammatory context and have similar effects than the ones described with IL-1ß. These results reinforce the idea that intraparietal amyloid deposits triggering adhesion molecules in endothelial cells, contribute to the transition of VSMCs to an inflammatory/de-differentiated phenotype. Therefore, we suggest that acute inflammatory episodes may increase vascular alterations and contribute to the ontogenesis of CAA.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/irrigación sanguínea , Desdiferenciación Celular , Inflamación/inmunología , Músculo Liso Vascular/citología , Músculo Liso Vascular/inmunología , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer , Animales , Células Cultivadas , Angiopatía Amiloide Cerebral/inmunología , Medios de Cultivo Condicionados , Activación Enzimática , Interleucina-1beta/farmacología , Ratones , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
17.
Arch Neurol ; 69(6): 773-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22351850

RESUMEN

BACKGROUND: In amyloid (Aß)-related angiitis (ABRA)of the central nervous system (CNS), cerebral amyloid angiopathy occurs in association with primary vasculitis of small- and medium-sized leptomeningeal and cortical arteries. It has been suggested that ABRA is triggered by vascular deposition of A followed by an Aß-directed (auto)immune response. OBJECTIVE: To provide a detailed description of the cellular composition of the inflammatory infiltrates in the cerebrospinal fluid (CSF) and CNS and their response to immunotherapy in a typical case of ABRA. DESIGN: Report of a single case. SETTING: Neurologic referral center. PATIENT: 67-year-old white woman. MAIN OUTCOME MEASURES: Neurologic examination,magnetic resonance imaging, lumbar puncture, flow cytometry,leptomeningeal biopsy, and histopathologic analysis. RESULTS: In a typical case of ABRA, we demonstrate for the first time the presence of a vast majority of partially activated CD4(+) T cells in CSF and leptomeningeal and parenchymal (peri)vascular infiltrates, which were frequently found in close proximity to major histocompatibility complex (MHC) class II-expressing microglia, epithelioid macrophages, and multinucleated giant cells containing intracellular deposits of Aß. CONCLUSION: Our findings support the notion of adaptive Aß-directed autoimmunity as the underlying pathogenic mechanism in ABRA.


Asunto(s)
Péptidos beta-Amiloides/líquido cefalorraquídeo , Linfocitos T CD4-Positivos/patología , Angiopatía Amiloide Cerebral , Vasculitis del Sistema Nervioso Central , Anciano , Angiopatía Amiloide Cerebral/líquido cefalorraquídeo , Angiopatía Amiloide Cerebral/complicaciones , Angiopatía Amiloide Cerebral/inmunología , Femenino , Citometría de Flujo , Humanos , Inmunoterapia , Imagen por Resonancia Magnética , Vasculitis del Sistema Nervioso Central/líquido cefalorraquídeo , Vasculitis del Sistema Nervioso Central/complicaciones , Vasculitis del Sistema Nervioso Central/inmunología
18.
J Neuroimmunol ; 244(1-2): 70-83, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22326143

RESUMEN

We evaluated the therapeutic efficacy of combined treatment of Aß-immunization with simvastatin in an Alzheimer mouse model at age 22 months. DNA prime-adenovirus boost immunization induced modest anti-Aß titers and simvastatin increased the seropositive rate. Aß-KLH was additionally administered to boost the titers. Irrespective of simvastatin, the immunization did not decrease cerebral Aß deposits but increased soluble Aß and tended to exacerbate amyloid angiopathy in the hippocampus. The immunization increased cerebral invasion of leukocytes and simvastatin counteracted the increase. Thus, modest anti-Aß titers can increase soluble Aß and simvastatin may reduce inflammation associated with vaccination in aged Alzheimer mouse models.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Simvastatina/uso terapéutico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Vacunas contra el Alzheimer/uso terapéutico , Animales , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/prevención & control , Modelos Animales de Enfermedad , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Neurobiol Aging ; 33(2): 432.e1-432.e13, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21371785

RESUMEN

Cerebrovascular amyloidosis is caused by amyloid accumulation in walls of blood vessel walls leading to hemorrhagic stroke and cognitive impairment. Transforming growth factor-ß1 (TGF-ß1) expression levels correlate with the degree of cerebrovascular amyloid deposition in Alzheimer's disease (AD) and TGF-ß1 immunoreactivity in such cases is increased along the cerebral blood vessels. Here we show that a nasally administered proteosome-based adjuvant activates macrophages and decreases vascular amyloid in TGF-ß1 mice. Animals were nasally treated with a proteosome-based adjuvant on a weekly basis for 3 months beginning at age 13 months. Using magnetic resonance imaging (MRI) we found that while control animals showed a significant cerebrovascular pathology, proteosome-based adjuvant prevents further brain damage and prevents pathological changes in the blood-brain barrier. Using an object recognition test and Y-maze, we found significant improvement in cognition in the treated group. Our findings support the potential use of a macrophage immunomodulator as a novel approach to reduce cerebrovascular amyloid, prevent microhemorrhage, and improve cognition.


Asunto(s)
Angiopatía Amiloide Cerebral/inmunología , Angiopatía Amiloide Cerebral/terapia , Cisteína Endopeptidasas/administración & dosificación , Modelos Animales de Enfermedad , Lipopolisacáridos/administración & dosificación , Activación de Macrófagos/efectos de los fármacos , Factor de Crecimiento Transformador beta1/inmunología , Administración por Inhalación , Animales , Combinación de Medicamentos , Inmunoterapia/métodos , Ratones , Ratones Transgénicos , Factor de Crecimiento Transformador beta1/genética , Resultado del Tratamiento
20.
Nanomedicine ; 8(2): 250-60, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21704598

RESUMEN

Alzheimer's disease amyloid ß (Aß) proteins accumulate in the cerebral vasculature and cause cerebral amyloid angiopathy (CAA). The objective of this study was to resolve critical formulation issues in developing nanoparticles (NPs) capable of permeating the blood brain barrier (BBB) and targeting cerebrovascular Aß proteins. To achieve this objective we designed immuno-nanovehicles, which are chitosan-coated poly lactic-co-glycolic acid (PLGA) NPs conjugated with a novel anti-Aß antibody. Measurements made according to Derjaguin-Landau-Verwey-Overbeek (DLVO) theory indicated that the immuno-nanovehicles have a much lower propensity to aggregate than the control nanovehicles. Immuno-nanovehicles showed enhanced uptake at the BBB and better targeting of the Aß proteins deposited in the CAA model in vitro in comparison with the control nanovehicles. In addition, chitosan enhanced aqueous dispersibility and increased the stability of immuno-nanovehicles during lyophilization, thus transforming them into ideal vehicles for delivering therapeutic and diagnostic agents to the cerebral vasculature ridden with vascular amyloid. FROM THE CLINICAL EDITOR: In this study, the authors report the development of chitosan-coated PLGA nanoparticles conjugated with anti-amyloid antibody to be used as immuno-nanovehicles to image cerebral amyloid angiopathy deposits in vivo. This method enables delivering therapeutic and diagnostic agents to the cerebral vasculature ridden with vascular amyloid.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Anticuerpos Antiidiotipos/inmunología , Angiopatía Amiloide Cerebral/diagnóstico , Quitosano/química , Nanopartículas/química , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Angiopatía Amiloide Cerebral/inmunología , Quitosano/metabolismo , Diagnóstico por Imagen/métodos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...