Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Nature ; 615(7953): 668-677, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890231

RESUMEN

Extracellular deposition of amyloid-ß as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-ß or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.


Asunto(s)
Encéfalo , Microglía , Ovillos Neurofibrilares , Linfocitos T , Tauopatías , Animales , Ratones , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Microglía/inmunología , Microglía/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/inmunología , Proteínas tau/metabolismo , Tauopatías/inmunología , Tauopatías/metabolismo , Tauopatías/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Placa Amiloide/inmunología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Innata
2.
Biochemistry ; 59(4): 341-342, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31944100

RESUMEN

The deposition of amyloid-ß (Aß) plaques and tau-based neurofibrillary tangles is a neuropathological feature of Alzheimer's disease (AD). While studies have shown that the Aß and tau interaction results in elevated AD pathology, the molecular linkage and mechanism of interaction of Aß and tau are unclear. A recent study demonstrated the direct interaction between the Aß core and specific regions of tau that facilitates pathological cross-seeding via a shared epitope. The data suggest that targeting the common epitope could be a more effective treatment strategy rather than targeting only Aß or only tau. The findings have an important clinical significance for AD and related tauopathies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Animales , Epítopos , Humanos , Ovillos Neurofibrilares/inmunología , Tauopatías/patología , Proteínas tau/inmunología
3.
Sci Rep ; 9(1): 15455, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31664089

RESUMEN

Pathological tau correlates well with cognitive impairments in Alzheimer's disease (AD) patients and therefore represents a promising target for immunotherapy. Targeting an appropriate B cell epitope in pathological tau could in theory produce an effective reduction of pathology without disrupting the function of normal native tau. Recent data demonstrate that the N-terminal region of tau (aa 2-18), termed the "phosphatase activation domain (PAD)", is hidden within native Tau in a 'paperclip'-like conformation. Conversely, PAD is exposed in pathological tau and plays an essential role in the inhibition of fast axonal transport and tau polymerization. Thus, we hypothesized that anti-tau2-18 antibodies may safely and specifically reduce pathological tau and prevent further aggregation, which in turn would neutralize tau toxicity. Therefore, we evaluated the immunogenicity and therapeutic efficacy of our MultiTEP platform-based vaccine targeting tau2-18 formulated with AdvaxCpG adjuvant (AV-1980R/A) in PS19 tau transgenic mice. The AV-1980R/A induced extremely high antibody responses and the resulting sera recognized neurofibrillary tangles and plaque-associated dystrophic neurites in AD brain sections. In addition, under non-denaturing conditions AV-1980R/A sera preferentially recognized AD-associated tau. Importantly, vaccination also prevented age-related motor and cognitive deficits in PS19 mice and significantly reduced insoluble total and phosphorylated tau species. Taken together, these findings suggest that predominantly targeting misfolded tau with AV-1980R/A could represent an effective strategy for AD immunotherapy.


Asunto(s)
Epítopos/inmunología , Monoéster Fosfórico Hidrolasas/metabolismo , Vacunas/inmunología , Proteínas tau/inmunología , Animales , Formación de Anticuerpos , Inmunoterapia , Ratones , Ovillos Neurofibrilares/inmunología , Fosforilación , Proteínas tau/química
4.
PLoS One ; 14(5): e0217216, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31120951

RESUMEN

Tauopathies represent a heterogeneous group of neurodegenerative disorders characterized by abnormal deposition of the hyperphosphorylated microtubule-associated protein tau. Chronic neuroinflammation in tauopathies is driven by glial cells that potentially trigger the disruption of the blood-brain barrier (BBB). Pro-inflammatory signaling molecules such as cytokines, chemokines and adhesion molecules produced by glial cells, neurons and endothelial cells, in general, cooperate to determine the integrity of BBB by influencing vascular permeability, enhancing migration of immune cells and altering transport systems. We considered the effect of tau about vascular permeability of peripheral blood cells in vitro and in vivo using primary rat BBB model and transgenic rat model expressing misfolded truncated protein tau. Immunohistochemistry, electron microscopy and transcriptomic analysis were employed to characterize the structural and functional changes in BBB manifested by neurofibrillary pathology in a transgenic model. Our results show that misfolded protein tau ultimately modifies the endothelial properties of BBB, facilitating blood-to-brain cell transmigration. Our results suggest that the increased diapedesis of peripheral cells across the BBB, in response to tau protein, could be mediated by the increased expression of endothelial signaling molecules, namely ICAM-1, VCAM-1, and selectins. We suggest that the compensation of BBB in the diseased brain represents a crucial factor in neurodegeneration of human tauopathies.


Asunto(s)
Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Ovillos Neurofibrilares/inmunología , Linfocitos T/inmunología , Tauopatías/inmunología , Proteínas tau/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuroglía/inmunología , Neuroglía/metabolismo , Neuroglía/patología , Ratas , Ratas Endogámicas SHR , Ratas Transgénicas , Linfocitos T/metabolismo , Linfocitos T/patología , Tauopatías/metabolismo , Tauopatías/patología
5.
Acta Neuropathol ; 133(5): 767-783, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28341999

RESUMEN

Several reports have described the presence of antibodies against Alzheimer's disease-associated hyperphosphorylated forms of tau in serum of healthy individuals. To characterize the specificities that can be found, we interrogated peripheral IgG+ memory B cells from asymptomatic blood donors for reactivity to a panel of phosphorylated tau peptides using a single-cell screening assay. Antibody sequences were recovered, cloned, and expressed as full-length IgGs. In total, 52 somatically mutated tau-binding antibodies were identified, corresponding to 35 unique clonal families. Forty-one of these antibodies recognize epitopes in the proline-rich and C-terminal domains, and binding of 26 of these antibodies is strictly phosphorylation dependent. Thirteen antibodies showed inhibitory activity in a P301S lysate seeded in vitro tau aggregation assay. Two such antibodies, CBTAU-7.1 and CBTAU-22.1, which bind to the proline-rich and C-terminal regions of tau, respectively, were characterized in more detail. CBTAU-7.1 recognizes an epitope that is similar to that of murine anti-PHF antibody AT8, but has different phospho requirements. Both CBTAU-7.1 and CBTAU-22.1 detect pathological tau deposits in post-mortem brain tissue. CBTAU-7.1 reveals a similar IHC distribution pattern as AT8, immunostaining (pre)tangles, threads, and neuritic plaques. CBTAU-22.1 shows selective detection of neurofibrillary changes by IHC. Taken together, these results suggest the presence of an ongoing antigen-driven immune response against tau in healthy individuals. The wide range of specificities to tau suggests that the human immune repertoire may contain antibodies that can serve as biomarkers or be exploited for therapy.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Epítopos/inmunología , Memoria Inmunológica/inmunología , Ovillos Neurofibrilares/inmunología , Proteínas tau/metabolismo , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos/fisiología , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Epítopos/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/patología , Fosforilación , Adulto Joven
6.
J Immunol ; 198(6): 2394-2402, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28188247

RESUMEN

The prevalence of neurodegenerative disease and arthritis increases with age. Despite both processes being associated with immune activation and inflammation, little is known about the mechanistic interactions between neurodegenerative disease and arthritis. In this article, we show that tau-transgenic (tau-tg) mice that develop neurodegenerative disease characterized by deposition of tau tangles in the brain are highly susceptible to developing arthritis. Already at steady-state conditions, tau-tg mice exhibit peripheral immune activation that is manifested by higher numbers of granulocytes, plasmablasts, and inflammatory Ly6Chi CCR2+ monocytes, as well as increased levels of proinflammatory cytokines, such as TNF-α and IL-17. Upon induction of collagen-induced arthritis (CIA), tau-tg mice displayed an increased incidence and an earlier onset of CIA that was associated with a more pronounced inflammatory cytokine response. Furthermore, induction of CIA led to significantly elevated numbers of Iba-1-expressing cells in the brain, indicative of microglia activation, and the formation of anti-tau Abs in tau-tg mice. These changes were accompanied by the resolution of tau tangles and significantly decreased neurodegenerative pathology. In summary, these data show that neurodegenerative disease enhances the development of arthritis. In addition, arthritis, once induced, triggers innate immune responses in the brain, leading to resolution of neurodegenerative changes.


Asunto(s)
Encéfalo/inmunología , Microglía/inmunología , Proteínas tau/metabolismo , Animales , Artritis Experimental , Autoanticuerpos/sangre , Proteínas de Unión al Calcio/metabolismo , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Enfermedades Neurodegenerativas , Ovillos Neurofibrilares/inmunología , Proteínas tau/genética , Proteínas tau/inmunología
7.
J Control Release ; 243: 1-10, 2016 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-27671875

RESUMEN

Detection of intracerebral targets with imaging probes is challenging due to the non-permissive nature of blood-brain barrier (BBB). The present work describes two novel single-domain antibodies (VHHs or nanobodies) that specifically recognize extracellular amyloid deposits and intracellular tau neurofibrillary tangles, the two core lesions of Alzheimer's disease (AD). Following intravenous administration in transgenic mouse models of AD, in vivo real-time two-photon microscopy showed gradual extravasation of the VHHs across the BBB, diffusion in the parenchyma and labeling of amyloid deposits and neurofibrillary tangles. Our results demonstrate that VHHs can be used as specific BBB-permeable probes for both extracellular and intracellular brain targets and suggest new avenues for therapeutic and diagnostic applications in neurology.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Ovillos Neurofibrilares/inmunología , Placa Amiloide/inmunología , Anticuerpos de Dominio Único/inmunología , Administración Intravenosa , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Microscopía/métodos , Anticuerpos de Dominio Único/metabolismo , Distribución Tisular
8.
J Pathol ; 235(5): 721-30, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25430817

RESUMEN

Amyloid ß peptide (Aß) immunization of Alzheimer's disease (AD) patients has been reported to induce amyloid plaque removal, but with little impact on cognitive decline. We have explored the consequences of Aß immunotherapy on neurons in post mortem brain tissue. Eleven immunized (AN1792, Elan Pharmaceuticals) AD patients were compared to 28 non-immunized AD cases. Immunohistochemistry on sections of neocortex was performed for neuron-specific nuclear antigen (NeuN), neurofilament protein (NFP) and phosphorylated-(p)PKR (pro-apoptotic kinase detected in degenerating neurons). Quantification was performed for pPKR and status spongiosis (neuropil degeneration), NeuN-positive neurons/field, curvature of the neuronal processes and interneuronal distance. Data were corrected for age, gender, duration of dementia and APOE genotype and also assessed in relation to Aß42 and tau pathology and key features of AD. In non-immunized patients, the degree of neuritic curvature correlated with spongiosis and pPKR, and overall the neurodegenerative markers correlated better with tau pathology than Aß42 load. Following immunization, spongiosis increased, interneuronal distance increased, while the number of NeuN-positive neurons decreased, consistent with enhanced neuronal loss. However, neuritic curvature was reduced and pPKR was associated with Aß removal in immunized patients. In AD, associations of spongiosis status, curvature ratio and pPKR load with microglial markers Iba1, CD68 and CD32 suggest a role for microglia in neurodegeneration. After immunization, correlations were detected between the number of NeuN-positive neurons and pPKR with Iba1, CD68 and CD64, suggesting that microglia are involved in the neuronal loss. Our findings suggest that in established AD this form of active Aß immunization may predominantly accelerate loss of damaged degenerating neurons. This interpretation is consistent with in vivo imaging indicating an increased rate of cerebral atrophy in immunized AD patients.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Vacunas contra el Alzheimer/uso terapéutico , Péptidos beta-Amiloides/uso terapéutico , Neocórtex/efectos de los fármacos , Neuronas/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/análisis , Antígenos Nucleares/análisis , Autopsia , Biomarcadores/análisis , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neocórtex/química , Neocórtex/inmunología , Neocórtex/patología , Degeneración Nerviosa , Proteínas del Tejido Nervioso/análisis , Ovillos Neurofibrilares/efectos de los fármacos , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/patología , Proteínas de Neurofilamentos/análisis , Neuronas/química , Neuronas/inmunología , Neuronas/patología , Fragmentos de Péptidos/análisis , Fosforilación , Placa Amiloide , Resultado del Tratamiento , eIF-2 Quinasa/análisis , Proteínas tau/análisis
9.
J Neurosci ; 34(12): 4260-72, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24647946

RESUMEN

Recent findings suggest that tau oligomers, which form before neurofibrillary tangles (NFTs), are the true neurotoxic tau entities in neurodegenerative tauopathies, including Alzheimer's disease (AD). Studies in animal models of tauopathy suggest that tau oligomers play a key role in eliciting behavioral and cognitive impairments. Here, we used a novel tau oligomer-specific monoclonal antibody (TOMA) for passive immunization in mice expressing mutant human tau. A single dose of TOMA administered either intravenously or intracerebroventricularly was sufficient to reverse both locomotor and memory deficits in a mouse model of tauopathy for 60 d, coincident with rapid reduction of tau oligomers but not phosphorylated NFTs or monomeric tau. Our data demonstrate that antibody protection is mediated by extracellular and rapid peripheral clearance. These findings provide the first direct evidence in support of a critical role for tau oligomers in disease progression and validate tau oligomers as a target for the treatment of AD and other neurodegenerative tauopathies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Inmunización Pasiva , Ovillos Neurofibrilares/inmunología , Tauopatías/terapia , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Fosforilación , Tauopatías/genética , Tauopatías/inmunología , Tauopatías/metabolismo , Proteínas tau/genética
10.
Exp Neurol ; 248: 451-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23876516

RESUMEN

The recent studies of others and of us showing robust efficacy of anti-tangle immunotherapy, directed against phosphorylated (phos)-tau protein, may pave the way to clinical trials of phos-tau immunotherapy in Alzheimer's-disease and other tauopathies. At this stage addressing the safety of the phos-tau-immunotherapy is highly needed, particularly since we have previously shown the neurotoxic potential of tau-immunotherapy, specifically of full-length unphosphorylated-tau vaccine under a CNS-proinflammatory milieu [induced by emulsification in complete-Freund's-adjuvant (CFA) and pertussis-toxin (PT)] in young wild-type (WT)-mice. The aim of our current study was to address safety aspects of the phos-tau-immunotherapy in both neurofibrillary-tangle (NFT)-mice as well as in WT-mice, under challenging conditions of repeated immunizations with phos-tau peptides under a CNS-proinflammatory milieu. NFT- and WT-mice were repeatedly immunized (7 injections in adult-, 4 in aged-mice) with phos-tau peptides emulsified in CFA-PT. A paralytic disease was evident in the phos-tau-immunized adult NFT-mice, developing progressively to 26.7% with the number of injections. Interestingly, the WT-mice were even more prone to develop neuroinflammation following phos-tau immunization, affecting 75% of the immunized mice. Aged mice were less prone to neuroinflammatory manifestations. Anti-phos-tau antibodies, detected in the serum of immunized mice, partially correlated with the neuroinflammation in WT-mice. This points that repeated phos-tau immunizations in the frame of a proinflammatory milieu may be encephalitogenic to tangle-mice, and more robustly to WT-mice, indicating that - under certain conditions - the safety of phos-tau immunotherapy is questionable.


Asunto(s)
Encefalitis/etiología , Inmunización/efectos adversos , Inmunoterapia/efectos adversos , Tauopatías/etiología , Proteínas tau/administración & dosificación , Proteínas tau/inmunología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Fosforilación , Tauopatías/inmunología , Tauopatías/metabolismo , Proteínas tau/metabolismo
11.
Curr Alzheimer Res ; 10(3): 217-28, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23534533

RESUMEN

The drawbacks of amyloid immunotherapy, including the development of encephalitis, the lack of clinical improvement and of any effect on neurofibrillary tangles (NFTs), coupled with the central role of NFTs in dementia, may point that clearance of amyloid pathology is not sufficient for improving the dementia symptoms in Alzheimer's disease (AD) patients. This further supported the concept that immunotherapy targeting the NFT proteinous aggregates may be preferential. Yet, the encephalitogenicity of full-length tau protein under a proinflammatory CNS milieu, reported by us in immunized mice, demands to carefully and selectively target pathological tau, while not the normal functional tau, and assuring both efficacy (anti-NFT effect) as well as safety (free of encephalitis) of a potential vaccine. Accumulating evidence from animal studies shows that tau-immunotherapy, targeting selectively pathological tau, particularly the phosphorylated-tau isoforms, reduces the tau-pathology and improves the symptoms of dementia. These findings are based on studies from different research groups, including our laboratory, conducted in different animal models and using various immunization protocols. There is also evidence that the decrease in NFTs is antibody-mediated involving the endosomal/ lysosomal pathway. No adverse effects were reported by the research groups, including also our study in which mice were immunized with a single injection of phosphorylated-tau peptide under a CNS proinflammatory milieu. In this review, I discuss the studies reported in this field, focusing on different approaches, different immunization protocols and mechanistic aspects, with a focus on the promising efficacy of the tau-immunotherapy, while addressing the safety issues already in the preclinical stage, before progressing to clinical trials.


Asunto(s)
Enfermedad de Alzheimer/terapia , Inmunoterapia/métodos , Tauopatías/terapia , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Humanos , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/patología
12.
J Neuroinflammation ; 10: 25, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23394121

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative dementia characterized by the decline of cognition and the presence of neuropathological changes including neuronal loss, neurofibrillary pathology and extracellular senile plaques. A neuroinflammatory process is also triggered and complement activation has been hypothesized to have a relevant role in this local inflammatory response. C5a, a proinflammatory anaphylatoxin generated after complement activation, exerts its chemotactic and inflammatory functions through the CD88 receptor while the more recently discovered C5L2 receptor has been postulated to have an anti-inflammatory role. Previously, we reported that a CD88 specific antagonist (PMX205) decreased the pathology and improved cognition in transgenic models of AD suggesting that C5a/C5aR interaction has an important role in the progression of the disease. METHODS: The present study characterizes the expression of the two receptors for C5a in human brain with confirmed post mortem diagnosis of vascular dementia (VD) or AD as well as age matched controls by immunohistochemistry and Western blot analysis using several antibodies against different epitopes of the human receptors. RESULTS: The CD88 and C5L2 antibodies revealed increased expression of both receptors in AD samples as compared to age-matched controls or VD brain tissue by Western blot and immunohistochemistry, using multiple antibodies and distinct cohorts of brain tissue. Immunostaining showed that both the C5L2 and CD88 antibodies similarly labeled abundant neurofibrillary tangles, neuropil threads and dystrophic neurites associated with plaques in the hippocampus and frontal cortex of AD cases. In contrast, little or no neuronal staining, tangles or dystrophic neurites associated with plaques were observed in control or VD brains. CD88 and C5L2 receptors are associated with both early (AT8) and mature (PHF1) neurofibrillary tangles and can be found either independently or colocalized with each other. CONCLUSIONS: The observed association of CD88 and C5L2 with neurofibrillary pathology suggests a common altered pathway of degradation.


Asunto(s)
Encéfalo/metabolismo , Ovillos Neurofibrilares/metabolismo , Receptores de Quimiocina/metabolismo , Receptores de Complemento/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/patología , Complemento C5a/biosíntesis , Complemento C5a/metabolismo , Demencia Vascular/metabolismo , Demencia Vascular/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/patología , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/biosíntesis , Receptores de Complemento/biosíntesis
13.
Neurochem Int ; 58(3): 301-20, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21167244

RESUMEN

Plaques and tangles are highly and significantly enriched in herpes simplex (HSV-1) binding proteins (by 11 and 15 fold respectively (P<4.47466E-39) and 132/341 (39%) of the known HSV-1 binding partners or associates are present in these structures. The classes involved include the majority (63-100%) of the known HSV-1 host protein carriers and receptors, 85-91% of the viral associated proteins involved in endocytosis, intracellular transport and exocytosis and 71% of the host proteins associated with the HSV-1 virion. The viral associated proteins found in plaques or tangles trace out a complete itinerary of the virus from entry to exocytosis and the virus also binds to plaque or tangle components involved in apoptosis, DNA transcription, translation initiation, protein chaperoning, the ubiquitin/proteasome system and the immune network. Along this route, the virus deletes mitochondrial DNA, as seen in Alzheimer's disease, sequesters the neuroprotective peptide, ADNP, and interferes with key proteins related to amyloid precursor protein processing and signalling as well as beta-amyloid processing, microtubule stability and tau phosphorylation, the core pathologies of Alzheimer's disease. Amyloid-containing plaques or neurofibrillary tangles also contain a large number of complement, acute phase and immune-related proteins, and the presence of these pathogen defence related classes along with HSV-1 binding proteins suggests that amyloid plaques and tangles represent cemeteries for a battle between the virus and the host's defence network. The presence of the complement membrane attack complex in Alzheimer's disease neurones suggests that complement mediated neuronal lysis may be a consequence of this struggle. HSV-1 infection is known to increase beta-amyloid deposition and tau phosphorylation and also results in cortical and hippocampal neuronal loss, cerebral shrinkage and memory deficits in mice. This survey supports the contention that herpes simplex viral infection contributes to Alzheimer's disease, in genetically predisposed individuals. Genetic conditioning effects are likely to be important, as all of the major risk promoting genes in Alzheimer's disease (apolipoprotein E, clusterin, complement receptor 1 and the phosphatidylinositol binding clathrin assembly protein PICALM), and many lesser susceptibility genes, are related to the herpes simplex life cycle. 33 susceptibility genes are related to the immune system. Vaccination or antiviral agents and immune suppressants should therefore perhaps be considered as viable therapeutic options, prior to, or in the early stages of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/virología , Proteínas del Sistema Complemento/fisiología , Encefalitis por Herpes Simple/complicaciones , Herpesvirus Humano 1/inmunología , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/patología , Neuronas/patología , Placa Amiloide/inmunología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/patología , Humanos , Ratones , Ovillos Neurofibrilares/virología , Neuronas/inmunología , Neuronas/virología , Placa Amiloide/patología , Placa Amiloide/virología
14.
J Neural Transm (Vienna) ; 117(8): 919-47, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20632195

RESUMEN

Generation of neurotoxic amyloid beta peptides and their deposition along with neurofibrillary tangle formation represent key pathological hallmarks in Alzheimer's disease (AD). Recent evidence suggests that inflammation may be a third important component which, once initiated in response to neurodegeneration or dysfunction, may actively contribute to disease progression and chronicity. Various neuroinflammatory mediators including complement activators and inhibitors, chemokines, cytokines, radical oxygen species and inflammatory enzyme systems are expressed and released by microglia, astrocytes and neurons in the AD brain. Degeneration of aminergic brain stem nuclei including the locus ceruleus and the nucleus basalis of Meynert may facilitate the occurrence of inflammation in their projection areas given the antiinflammatory and neuroprotective action of their key transmitters norepinephrine and acetylcholine. While inflammation has been thought to arise secondary to degeneration, recent experiments demonstrated that inflammatory mediators may stimulate amyloid precursor protein processing by various means and therefore can establish a vicious cycle. Despite the fact that some aspects of inflammation may even be protective for bystander neurons, antiinflammatory treatment strategies should therefore be considered. Non-steroidal anti-inflammatory drugs have been shown to reduce the risk and delay the onset to develop AD. While, the precise molecular mechanism underlying this effect is still unknown, a number of possible mechanisms including cyclooxygenase 2 or gamma-secretase inhibition and activation of the peroxisome proliferator activated receptor gamma may alone or, more likely, in concert account for the epidemiologically observed protection.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo/patología , Inflamación/etiología , Inflamación/patología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/inmunología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/inmunología , Cromogranina A/inmunología , Cromogranina A/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/uso terapéutico , Ovillos Neurofibrilares/inmunología
15.
Exp Neurol ; 224(2): 472-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20546729

RESUMEN

As an abnormally folded and aggregated protein, tau composed of neurofibrillary tangles (NFTs) in Alzheimer's disease and other tauopathies seems to be a candidate for immunotherapy. Yet, the encephalitogenicity of full-length tau protein, recently reported by us in immunized mice, demands to carefully and selectively target pathological tau and address both efficacy (anti-NFT effect) and safety (free of encephalitis). We immunized NFT mice with NFT-related phosphorylated (phos) tau peptides, using an immunization protocol aimed to predispose a proinflammatory milieu in CNS as a set up to detect biohazard, an approach we used when the neurotoxicity of full-length tau was detected [use of complete Freund adjuvant (CFA) with pertussis toxin (PT)]. A decrease of about 40% in NFT burden in CNS was demonstrated and was accompanied with an increase in microglial burden. Anti-phos-tau antibodies were detected in serum and blood vessels in the CNS, while no encephalitogenicity (free of clinical neurological deficits, of adverse effects on brain inflammatory cells and of axonal damage) was recorded. The level of the lysosomal proteases, cathepsins D and L, was affected in the immunized mice suggesting the possible involvement of the lysosomal system in the decrease of NFTs. The robust anti-NFT effect and the lack of encephalitogenicity in NFT mice immunized with phos-tau peptides, even though CFA with PT was included in vaccine, point to their anti-NFT therapeutic potential.


Asunto(s)
Ovillos Neurofibrilares/patología , Fragmentos de Péptidos/inmunología , Fosfoproteínas/uso terapéutico , Tauopatías/terapia , Proteínas tau/uso terapéutico , Animales , Anticuerpos/sangre , Astrocitos/inmunología , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Catepsina D/metabolismo , Catepsina L/metabolismo , Encefalitis/inducido químicamente , Femenino , Inmunización , Lisosomas/enzimología , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Ovillos Neurofibrilares/inmunología , Neuronas/inmunología , Neuronas/patología , Fragmentos de Péptidos/efectos adversos , Péptido Hidrolasas/metabolismo , Fosfoproteínas/efectos adversos , Tauopatías/inmunología , Tauopatías/patología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Proteínas tau/efectos adversos
16.
Acta Neuropathol ; 120(1): 13-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20532897

RESUMEN

Alzheimer's disease (AD) pathology is characterised by aggregation in the brain of amyloid-beta (Abeta) peptide and hyperphosphorylated tau (phospho-tau), although how these proteins interact in disease pathogenesis is unclear. Abeta immunisation results in removal of Abeta from the brain but cognitive decline continues to progress, possibly due to persistent phospho-tau. We quantified phospho-tau and Abeta42 in the brains of 10 AD patients (iAD) who were actively immunised with Abeta42 (AN1792, Elan Pharmaceuticals) compared with 28 unimmunised AD cases (cAD). The phospho-tau load was lower in the iAD than the cAD group in the cerebral cortex (cAD 1.08% vs. iAD 0.72%, P = 0.048), CA1 hippocampus (cAD 2.26% vs. iAD 1.05%; P = 0.001), subiculum (cAD 1.60% vs. iAD 0.31%; P = 0.001) and entorhinal cortex (cAD 1.10% vs. iAD 0.18%; P < 0.001). Assessment of the localisation within neurons of phospho-tau indicated that the Abeta immunotherapy-associated reduction was confined to neuronal processes, i.e. neuropil threads and dystrophic neurites. However, the phospho-tau accumulation in the neuronal cell bodies, contributing to neurofibrillary tangles, appeared not to be affected. In showing that Abeta immunisation can influence phospho-tau pathology, we confirm the position of Abeta as a target for modifying tau accumulation in AD and demonstrate a link between these proteins. However, the continuing progression of cognitive decline in AD patients after Abeta immunisation may be explained by its lack of apparent effect on tangles.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/inmunología , Neuronas/inmunología , Neuronas/metabolismo , Fragmentos de Péptidos/inmunología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Región CA1 Hipocampal/inmunología , Región CA1 Hipocampal/metabolismo , Corteza Entorrinal/inmunología , Corteza Entorrinal/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Inmunización , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neocórtex/inmunología , Neocórtex/metabolismo , Neuritas/inmunología , Neuritas/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Fosforilación , Resultado del Tratamiento
17.
Rev. chil. neuro-psiquiatr ; 48(1): 49-57, mar. 2010. ilus
Artículo en Español | LILACS | ID: lil-577345

RESUMEN

The progressive increase in life expectancy of the world population has fostered a major concern in order to find effective avenues for diagnosis of treatment of Alzheimer's disease (AD). Even tough AD pathogenesis is still unclear, new advances have allowed to understand that exposure of individuals to a series of environmental risk factors, named to as damage signals, play a main role in triggering the disease. This is important for AD prevention but also for the search of new treatment approaches. Activation of innate immunity in the central nervous system (CNS), essentially microglial cells, appears to be a key element in the neurodegenerative pathway As a matter of fact, when microglia cells are exposed continuously to damage signals such as metabolites from conditions of hyperlipidemia, hyperglycemia, oxidative stress, head injury and trauma, recurrent infections, in addition to supramolecular aggregates such as tau filaments or b-amyloid oligomers, among other anomalous protein filaments, they respond by triggering the inflammatory cascade. On this basis, we have postulated the neuroimmunomodulation hypothesis for Alzheimer's Disease. Therefore, we postulates that a long-term activation of brain innate immunity by a converging set of damage signals constitute a unifying mechanism that triggers the inflammatory cascade, thus leading to irreversible alteration in the neuronal cytoskeleton. These concerted alterations in signaling mechanisms will lead in neuronal cells to a final common pathway, tau hyperphosphorylations, with the consequent self-aggregation of modified tau and formation of paired helical filaments (PHFs), as the main triggering event for neurodegenration in AD.


El constante aumento en la expectativa de vida en la población mundial ha incrementado la preocupación hacia la búsqueda de la comprensión de la Enfermedad de Alzheimer (EA), así como de su diagnóstico temprano y tratamiento. Actualmente la etiopatogenia que conduce al desarrollo de la EA es aún difusa, pero se ha llegado a comprender que la exposición a una serie de distintos factores de riesgo, o señales de daño, está asociada al desencadenamiento de la EA. Esto es muy importante no solo para la prevención de esta devastadora enfermedad sino también para la búsqueda de avenidas efectivas para su tratamiento. En efecto, la activación de la inmunidad innata en el sistema nervioso central (SNC), esencialmente por las células microgliales, son un elemento clave en el proceso neurodegenerativo, cuando éstas son expuestas por períodos prolongados a señales de daño. Entre éstas están la hiperlipidemia, hiperglicemia, estrés oxidativo, traumatismos, infecciones recurrentes, oligomeros de -amiloide, agregados de tau, entre otros factores, los que desencadenarían una respuesta pro-inflamatoria persistente que conduce a la cascada neurodegenerativa. En base a esto, hemos postulado la teoría de la neuroinmunomodulación en la EA, y proponemos que la activación a largo plazo del sistema inmune innato por un conjunto de señales de daño constituye un mecanismo unificado que gatillo, una cascada inflamatoria que conduce a alteraciones irreversibles en el citoesqueleto. Estos mecanismos anómalos de señalización molecular llevarían a una vía final común que es la hiperfosforilación de la proteína tau, su autoagregación y formación de los PHFs, como desencadenantes claves en la neurodegeneración y desarrollo de la EA.


Asunto(s)
Humanos , Enfermedad de Alzheimer/inmunología , Inflamación/inmunología , Sistema Nervioso Central/inmunología , Citocinas , Inmunidad Innata/inmunología , Microglía/inmunología , Neuroinmunomodulación , Ovillos Neurofibrilares/inmunología , Factores Desencadenantes , Factores de Riesgo , Proteínas tau
18.
Curr Alzheimer Res ; 6(5): 446-50, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19874269

RESUMEN

Immunotherapies targeting the amyloid-beta (Abeta) peptide in Alzheimer's disease (AD) have consistently been effective in mouse studies and shown promise in clinical trials, although some setbacks have occurred. First, encephalitis was observed in a small subset of patients. More recent autopsy data from a few subjects suggests that clearance of Abeta plaques may not halt cognitive deterioration once impairments are evident, emphasizing the need for other more effective approaches at that stage of the disease. Another important target in AD is the neurofibrillary tangles and its precursors, composed primarily of hyperphosphorylated tau proteins, which correlate well with the degree of dementia. As Abeta and tau pathologies are likely synergistic, targeting both together may be more effective, and perhaps essential as early diagnosis prior to cognitive decline is currently unavailable. Also, Abeta immunotherapy results in a very limited indirect clearance of tau aggregates, showing the importance of developing a separate therapy that directly targets pathological tau. Our findings in two tangle mouse models indicate that active immunization targeting an AD phospho-tau epitope reduces aggregated tau in the brain and prevents/slows progression of the tangle-related behavioral phenotype, including cognitive impairment. These antibodies enter the brain and bind to pathological tau within neurons although the therapeutic effect may at least in part be due to clearance of extracellular tau that may have biological effects. We are currently clarifying the mechanism of these promising findings, determining its epitope specificity as well as assessing the feasibility of this approach for clinical trials.


Asunto(s)
Enfermedad de Alzheimer/terapia , Proteínas tau/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Humanos , Inmunoterapia , Ratones , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/patología , Fosforilación/inmunología
19.
Brain Res ; 1231: 132-42, 2008 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-18606155

RESUMEN

Neuritic dystrophy with amyloid burden and neurofibrillary tangles are pathological hallmarks of Alzheimer's disease. Genetic disruption of CD40 or CD40L alleviates amyloid burden, astrocytosis, and microgliosis in transgenic animal models of Alzheimer's disease. It has been reported that phosphorylated tau-positive dystrophic neurites are observed in transgenic mice over-expressing human mutant beta-amyloid precursor protein (Tg2576). Here, we studied the pattern of phosphorylated tau (labeled with AT8, CP13, PG5, and PHF1 antibodies) and plaques using immunohistochemical techniques. Phosphorylated tau-positive dystrophic neurites were exclusively associated with Congo red-positive plaques as previously reported. Further, we show that CD40L or CD40 deficiency reduces the mean ratio of dystrophic neurite area to congophilic plaque area and the level of expression of cdk5 and p35/p25 in mice. In addition, we show that in a human neuroblastoma cell line treated with CD40L, cdk5 and p35/p25 are increased. Together, our data suggest that CD40-CD40L interaction has an effect on tau phosphorylation independent of beta-amyloid pathology, and that this effect may occur through a decrease of cdk5 and p35/p25.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Placa Amiloide/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/fisiopatología , Antígenos CD40/efectos de los fármacos , Antígenos CD40/inmunología , Ligando de CD40/inmunología , Ligando de CD40/farmacología , Línea Celular Tumoral , Homólogo de la Proteína Chromobox 5 , Colorantes , Rojo Congo , Quinasa 5 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuritas/efectos de los fármacos , Neuritas/inmunología , Neuritas/metabolismo , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Fosforilación/efectos de los fármacos , Fosfotransferasas/efectos de los fármacos , Fosfotransferasas/metabolismo , Placa Amiloide/genética , Placa Amiloide/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA