Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 38(6): 1207-1214, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29948552

RESUMO

Despite years of research, Alzheimer's disease (AD) remains incurable and thus poses a major health challenge in coming years. This neurodegenerative disease belongs to a heterogeneous group of human tauopathies, characterized by the extracellular deposition of beta amyloid-Aß and intracellular accumulation of tau protein in neuronal and glial cells, whereby tau pathology best correlates with disease progression. For decades, several disease-modifying agents were brought to clinical studies with promising efficacy in preclinical trials; however, all of the subsequent clinical trials failed. Therefore, the pursuit for therapeutic agents for the treatment of AD and other tauopathies still continue. Recent evidences show previously unidentified role of peripheral immune system in regulating the inflammatory status of the brain, mainly the dendritic cells. A decrease in functionality and count of dendritic cells has been observed in Alzheimer's disease. Here, we discuss a potential role of dendritic cell-based vaccines as therapeutic approach in ameliorating disease pathogenesis in AD and other tauopathies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Encéfalo/metabolismo , Células Dendríticas/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/efeitos dos fármacos , Humanos , Neurônios/metabolismo , Tauopatias/tratamento farmacológico , Proteínas tau/metabolismo
2.
Cell Mol Neurobiol ; 38(1): 243-259, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28405903

RESUMO

Alzheimer's disease (AD) is a multifactorial disorder; neurofibrillary pathology composed of tau protein is found side by side with amyloid-ß deposits and extensive neuroinflammation. The immune system of the brain is considered as one of the factors that could influence the speed of the progression of AD neuropathology as a potential mediator of the damage induced by AD protein deposits. Alzheimer's disease pathology can be impacted by psychological stress; however, signalling pathways in background are not well known. We have explored possible avenues of how stress could influence the brain's immune system in a rat model of AD. Animals were subjected either to a single or multiple instances of immobilization stress. The analysis of a panel of immunity-related genes was used to evaluate the impact of stress on the immune response in the brain. We have identified 19 stress-responsive genes that are involved in neuroinflammation accompanying tau pathology: Nos2, Ptgs2, IL-8rb, C5, Mmp9, Cx3cr1, CD40lg, Adrb2, IL-6, IL-6r, IL-1r2, Ccl2, Ccl3, Ccl4, Ccl12, TNF-α, IL-1α, IL-1ß, IL-10. Most of them are deregulated under the stress conditions also in control animals; however, the magnitude of the response to either acute or chronic stress differs. This can lead to serious influence, most probably to acceleration of neurodegenerative phenotype in diseased animals. Several of the genes (IL-1ß, Casp1, Cx3cr1 and C5) are deregulated solely in tauopathic animals. The stress-induced changes in the inflammatory picture of the brain highlight the fact that the brain's immune response is highly responsive to environmental stimuli. The pattern of changes is indicative of an attempt to protect the brain in the short term, while being potentially detrimental to the response against a long-term pathological process such as neurofibrillary degeneration.


Assuntos
Encéfalo/imunologia , Imunidade Celular/fisiologia , Doenças Neurodegenerativas/imunologia , Estresse Psicológico/imunologia , Proteínas tau/imunologia , Animais , Encéfalo/metabolismo , Feminino , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/psicologia , Ratos , Ratos Endogâmicos SHR , Ratos Transgênicos , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Proteínas tau/metabolismo
3.
Biochim Biophys Acta ; 1852(7): 1219-29, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25772164

RESUMO

Accumulation of misfolded forms of microtubule associated, neuronal protein tau causes neurofibrillary degeneration typical of Alzheimer's disease and other tauopathies. This process is accompanied by elevated cellular stress and concomitant deregulation of heat-shock proteins. We used a transgenic rat model of tauopathy to study involvement of heat shock protein 27 (Hsp27) in the process of neurofibrillary degeneration, its cell type specific expression and correlation with the amount of insoluble tau protein aggregates. The expression of Hsp27-mRNA is more than doubled and levels of Hsp27 protein tripled in aged transgenic animals with tau pathology. The data revealed a strong positive and highly significant correlation between Hsp27-mRNA and amount of sarkosyl insoluble tau. Interestingly, intracellular accumulation of insoluble misfolded tau protein in neurons was associated with overexpression of Hsp27 almost exclusively in reactive astrocytes, not in neurons. The topological dissociation of neuronally expressed pathological tau and the induction of astrocytic Hsp27, GFAP, and Vimentin along with up-regulation of microglia specific markers such as CD18, CD68 and C3 point to cooperation of astrocytes, microglia and neurons in response to intra-neuronal accumulation of insoluble tau. Our data suggest that over expression of Hsp27 represents a part of microglia-mediated astrocytic response mechanism in the process of neurofibrillary degeneration, which is not necessarily associated with neuroprotection and which in contrary may accelerate neurodegeneration in late stage of the disease. This phenomenon should be considered during development of disease modifying strategies for treatment of tauopathies and AD via regulation of activity of Hsp27.


Assuntos
Astrócitos/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Neurônios/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Células Cultivadas , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Choque Térmico HSP27/genética , Humanos , Dobramento de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Vimentina/genética , Vimentina/metabolismo , Proteínas tau/química
4.
J Neuroinflammation ; 13: 15, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26792515

RESUMO

BACKGROUND: Brain norepinephrine (NE) plays an important role in the modulation of stress response and neuroinflammation. Recent studies indicate that in Alzheimer's disease (AD), the tau neuropathology begins in the locus coeruleus (LC) which is the main source of brain NE. Therefore, we investigated the changes in brain NE system and also the immune status under basal and stress conditions in transgenic rats over-expressing the human truncated tau protein. METHODS: Brainstem catecholaminergic cell groups (LC, A1, and A2) and forebrain subcortical (nucleus basalis of Meynert), hippocampal (cornu ammonis, dentate gyrus), and neocortical areas (frontal and temporal association cortices) were analyzed for NE and interleukin 6 (IL-6) mRNA levels in unstressed rats and also in rats exposed to single or repeated immobilization. Moreover, gene expression of NE-biosynthetic enzyme, tyrosine hydroxylase (TH), and several pro- and anti-inflammatory mediators were determined in the LC. RESULTS: It was found that tauopathy reduced basal NE levels in forebrain areas, while the gene expression of IL-6 was increased in all selected areas at the same time. The differences between wild-type and transgenic rats in brain NE and IL-6 mRNA levels were observed in stressed animals as well. Tauopathy increased also the gene expression of TH in the LC. In addition, the LC exhibited exaggerated expression of pro- and anti-inflammatory mediators (IL-6, TNFα, inducible nitric oxide synthases 2 (iNOS2), and interleukin 10 (IL-10)) in transgenic rats suggesting that tauopathy affects also the immune background in LC. Positive correlation between NE and IL-6 mRNA levels in cornu ammonis in stressed transgenic animals indicated the reduction of anti-inflammatory effect of NE. CONCLUSIONS: Our data thus showed that tauopathy alters the functions of LC further leading to the reduction of NE levels and exaggeration of neuroinflammation in forebrain. These findings support the assumption that tau-related dysfunction of LC activates the vicious circle perpetuating neurodegeneration leading to the development of AD.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Encefalite/etiologia , Norepinefrina/metabolismo , Tauopatias , Análise de Variância , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Citocinas/genética , Citocinas/metabolismo , Expressão Gênica/genética , Humanos , Masculino , Microdissecção , Óxido Nítrico Sintase Tipo II/metabolismo , Norepinefrina/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Transgênicos , Tauopatias/complicações , Tauopatias/genética , Tauopatias/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
5.
Stress ; 19(4): 395-405, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27484105

RESUMO

Neuroendocrine and behavioral stress responses are orchestrated by corticotropin-releasing hormone (CRH) and norepinephrine (NE) synthesizing neurons. Recent findings indicate that stress may promote development of neurofibrillary pathology in Alzheimer's disease. Therefore, we investigated relationships among stress, tau protein phosphorylation, and brain NE using wild-type (WT) and CRH-knockout (CRH KO) mice. We assessed expression of phosphorylated tau (p-tau) at the PHF-1 epitope and NE concentrations in the locus coeruleus (LC), A1/C1 and A2/C2 catecholaminergic cell groups, hippocampus, amygdala, nucleus basalis magnocellularis, and frontal cortex of unstressed, singly stressed or repeatedly stressed mice. Moreover, gene expression and protein levels of tyrosine hydroxylase (TH) and CRH receptor mRNA were determined in the LC. Plasma corticosterone levels were also measured. Exposure to a single stress increases tau phosphorylation throughout the brain in WT mice when compared to singly stressed CRH KO animals. In contrast, repeatedly stressed CRH KO mice showed exaggerated tau phosphorylation relative to WT controls. We also observed differences in extent of tau phosphorylation between investigated structures, e.g. the LC and hippocampus. Moreover, CRH deficiency leads to different responses to stress in gene expression of TH, NE concentrations, CRH receptor mRNA, and plasma corticosterone levels. Our data indicate that CRH effects on tau phosphorylation are dependent on whether stress is single or repeated, and differs between brain regions. Our findings indicate that CRH attenuates mechanisms responsible for development of stress-induced tau neuropathology, particularly in conditions of chronic stress. However, the involvement of central catecholaminergic neurons in these mechanisms remains unclear and is in need of further investigation.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Neurônios/metabolismo , Estresse Psicológico/metabolismo , Proteínas tau/metabolismo , Animais , Hormônio Liberador da Corticotropina/genética , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Fosforilação , RNA Mensageiro/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Restrição Física
6.
J Neural Transm (Vienna) ; 123(12): 1423-1433, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27287334

RESUMO

Huntington's disease is an incurable, adult-onset, autosomal dominant inherited disorder caused by an expanded trinucleotide repeat (CAG). In this study, we describe a Huntington's disease patient displaying clinical symptoms of the behavioural variant of frontotemporal dementia in the absence of tremor and ataxia. The clinical onset was at the age of 36 years and the disease progressed slowly (18 years). Genetic testing revealed expanded trinucleotide CAG repeats in the Huntingtin gene, together with a Glu318Gly polymorphism in presenilin 1. Neuropathological assessment revealed extensive amyloid ß (Aß) aggregates in all cortical regions. No inclusions displaying hyperphosphorylated tau or phosphorylated transactive response DNA-binding protein 43 (TDP43) were found. A high number of p62 (sequestosome 1) immunopositive intranuclear inclusions were seen mainly in the cortex, while subcortical areas were affected to a lesser extent. Confocal microscopy revealed that the majority of p62 intranuclear lesions co-localised with the fused-in-sarcoma protein (FUS) immunostaining. The morphology of the inclusions resembled intranuclear aggregates in Huntington's disease. The presented proband suffered from Huntington's disease showed atypical distribution of FUS positive intranuclear aggregates in the cortical areas with concomitant Alzheimer's disease pathology.


Assuntos
Encéfalo/metabolismo , Demência Frontotemporal/complicações , Doença de Huntington/complicações , Adulto , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Saúde da Família , Feminino , Compostos de Anéis Fundidos/metabolismo , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Presenilina-1/genética , Proteínas de Ligação a RNA/metabolismo , Expansão das Repetições de Trinucleotídeos/genética
7.
Eur J Neurosci ; 42(2): 1872-86, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25994480

RESUMO

Brainstem catecholaminergic neurons significantly participate in the regulation of neuroendocrine system activity, particularly during stressful conditions. However, so far the precise quantitative characterisation of basal and stress-induced changes in gene expression and protein levels of catecholaminergic biosynthetic enzymes in these neurons has been missing. Using a quantitative reverse transcription-polymerase chain reaction method, we investigated gene expression of catecholamine biosynthetic enzymes in brainstem noradrenergic and adrenergic cell groups in rats under resting conditions as well as in acutely and repeatedly stressed animals. For the first time, we described quantitative differences in basal levels of catecholamine biosynthetic enzyme mRNA in brainstem catecholaminergic ascending and descending projecting cell groups. Moreover, we found and defined some differences among catecholaminergic cell groups in the time-course of mRNA levels of catecholaminergic enzymes following a single and especially repeated immobilisation stress. The data obtained support the assumption that brainstem catecholaminergic cell groups represent a functionally differentiated system, which is highly (but specifically) activated in rats exposed to stress. Therefore, potential interventions for the treatment of stress-related diseases need to affect the activity of brainstem catecholaminergic neurons not uniformly but with some degree of selectivity.


Assuntos
Tronco Encefálico/metabolismo , Catecolaminas/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Resposta de Imobilidade Tônica/fisiologia , Feniletanolamina N-Metiltransferase/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Dopamina beta-Hidroxilase/genética , Masculino , Microdissecção , Feniletanolamina N-Metiltransferase/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/genética
8.
Gen Physiol Biophys ; 34(4): 337-52, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26221742

RESUMO

Impairment of "protein quality control" in neurons is associated with etiopathogenesis of neurodegenerative diseases. The worn-out products of cell metabolism should be safely eliminated via the proteasome, autophago-lysosome and exocytosis. Insufficient activity of these degradation mechanisms within neurons leads to the accumulation of toxic protein oligomers, which represent a starting material for development of neurodegenerative proteinopathy. The spectrum of CNS linked proteinopathies is particularly broad and includes Alzheimer's disease (AD), Parkinson's disease (PD), Lewy body dementia, Pick disease, Frontotemporal dementia, Huntington disease, Amyotrophic lateral sclerosis and many others. Although the primary events in etiopathogenesis of sporadic forms of these diseases are still unknown, it is clear that aging, in connection with decreased activity of ubiquitin proteasome system, is the most significant risk factor. In this review we discuss the pathogenic role and intracellular fate of the candidate molecules associated with onset and progression of AD and PD, the protein tau and α-synuclein in context with the function of ubiquitin proteasome system. We also discuss the possibility whether or not the strategies focused to re-establishment of neuroproteostasis via accelerated clearance of damaged proteins in proteasome could be a promising therapeutic approach for treatment of major neurodegenerative diseases.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitinas/metabolismo , Animais , Humanos , Modelos Neurológicos , Terapia de Alvo Molecular/métodos , Doenças Neurodegenerativas/terapia , Fármacos Neuroprotetores/uso terapêutico
9.
J Neuroinflammation ; 11: 161, 2014 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-25217135

RESUMO

BACKGROUND: Abnormal misfolded tau protein is a driving force of neurofibrillary degeneration in Alzheimer's disease. It has been shown that tau oligomers play a crucial role in the formation of intracellular neurofibrillary tangles. They are intermediates between soluble tau monomers and insoluble tau filaments and are suspected contributors to disease pathogenesis. Oligomeric tau can be released into the extracellular space and spread throughout the brain. This finding opens the question of whether brain macrophages or blood monocytes have the potential to phagocytose extracellular oligomeric tau. METHODS: We have used stable rat primary microglial cells, rat peripheral monocytes-derived macrophages, BV2 microglial and TIB67 macrophage immortalized cell lines that were challenged by tau oligomers prepared by an in vitro aggregation reaction. The efficiency of cells to phagocytose oligomeric protein was evaluated with confocal microscopy. The ability to degrade tau protein was analyzed by immunoblotting. RESULTS: Confocal microscopy analyses showed that macrophages were significantly more efficient in phagocytosing oligomerized tau proteins than microglial cells. In contrast to macrophages, microglia are able to degrade the internalized oligomeric tau only after stimulation with lipopolysaccharide (LPS). CONCLUSIONS: Our data suggests that microglia may not be the principal phagocytic cells able to target extracellular oligomeric tau. We found that peripheral macrophages display a high potency for elimination of oligomeric tau and therefore could play an important role in the modulation of neurofibrillary pathology in Alzheimer's disease.


Assuntos
Macrófagos/metabolismo , Microglia/metabolismo , Fagocitose/fisiologia , Proteínas tau/metabolismo , Animais , Western Blotting , Células Cultivadas , Espaço Extracelular/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Multimerização Proteica , Ratos , Ratos Sprague-Dawley
10.
J Immunol ; 187(5): 2732-9, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21813771

RESUMO

Neuroinflammation plays a key role in the pathogenesis of Alzheimer's disease and related tauopathies. We have previously shown that expression of nonmutated human truncated τ (151-391, 4R), derived from sporadic Alzheimer's disease, induced neurofibrillary degeneration accompanied by microglial and astroglial activation in the brain of transgenic rats. The aim of the current study was to determine the molecular mechanism underlying innate immune response induced by misfolded truncated τ. We found that purified recombinant truncated τ induced morphological transformation of microglia from resting into the reactive phenotype. Simultaneously, truncated τ caused the release of NO, proinflammatory cytokines (IL-1ß, IL-6, TNF-α), and tissue inhibitor of metalloproteinase-1 from the mixed glial cultures. Notably, when the pure microglial culture was activated with truncated τ, it displayed significantly higher levels of the proinflammatory cytokines, suggesting a key role of microglia in the τ-mediated inflammatory response. Molecular analysis showed that truncated τ increased the mRNA levels of three MAPKs (JNK, ERK1, p38ß) and transcription factors AP-1 and NF-κB that ultimately resulted in enhanced mRNA expression of IL-1ß, IL-6, TNF-α, and NO. Our results showed for the first time, to our knowledge, that misfolded truncated protein τ is able to induce innate immune response via a MAPK pathway. Consequently, we suggest that misfolded truncated protein τ represents a viable target for immunotherapy of Alzheimer's disease.


Assuntos
Imunidade Inata , Microglia/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Proteínas tau/imunologia , Animais , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Ratos , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
11.
J Neuroinflammation ; 9: 47, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22397366

RESUMO

Neurodegeneration, induced by misfolded tau protein, and neuroinflammation, driven by glial cells, represent the salient features of Alzheimer's disease (AD) and related human tauopathies. While tau neurodegeneration significantly correlates with disease progression, brain inflammation seems to be an important factor in regulating the resistance or susceptibility to AD neurodegeneration. Previously, it has been shown that there is a reciprocal relationship between the local inflammatory response and neurofibrillary lesions. Numerous independent studies have reported that inflammatory responses may contribute to the development of tau pathology and thus accelerate the course of disease. It has been shown that various cytokines can significantly affect the functional and structural properties of intracellular tau. Notwithstanding, anti-inflammatory approaches have not unequivocally demonstrated that inhibition of the brain immune response can lead to reduction of neurofibrillary lesions. On the other hand, our recent data show that misfolded tau could represent a trigger for microglial activation, suggesting the dual role of misfolded tau in the Alzheimer's disease inflammatory cascade. On the basis of current knowledge, we can conclude that misfolded tau is located at the crossroad of the neurodegenerative and neuroinflammatory pathways. Thus disease-modified tau represents an important target for potential therapeutic strategies for patients with Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encefalite/etiologia , Doenças Neurodegenerativas/etiologia , Deficiências na Proteostase/complicações , Proteínas tau/metabolismo , Animais , Encefalite/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Dobramento de Proteína
12.
Biochem Soc Trans ; 40(4): 681-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817716

RESUMO

Pathological truncations of human brain proteins represent the common feature of many neurodegenerative disorders including AD (Alzheimer's disease), Parkinson's disease and Huntington's disease. Protein truncations significantly change the structure and function of these proteins and thus can engender their pathological metamorphosis. We have shown previously that truncated forms of tau protein are contained in the core of the paired helical filaments that represent the main constituent of neurofibrillary pathology. Recently, we have identified truncated tau species of a different molecular signature. We have found that tau truncation is not produced by a random process, but rather by highly specific proteolytic cleavage and/or non-enzymatic fragmentation. In order to characterize the pathophysiology of AD-specific truncated tau species, we have used a transgenic rat model for AD expressing human truncated tau. Expression of the tau protein induces the formation of novel truncated tau species that originate from both transgenic human tau and endogenous rat tau proteins. Moreover, these truncated tau proteins are found exclusively in the misfolded fraction of tau, suggesting that they actively participate in the tau misfolding process. These findings corroborate further the idea that the appearance of truncated tau species starts a self-perpetuating cycle of further tau protein truncation leading to and accelerating tau misfolding and formation of neurofibrillary pathology.


Assuntos
Proteômica/métodos , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo
13.
Cell Mol Neurobiol ; 32(5): 837-45, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22222439

RESUMO

Tau protein misfolding is a pathological mechanism, which plays a critical role in the etiopathogenesis of neurodegeneration. However, it is not entirely known what kind of stimuli can induce the misfolding. It is believed that physical and emotional stresses belong to such risk factors. Although the influence of stress on the onset and progression of Alzheimer's disease (AD) has already been proposed, the molecular links between stresses and AD are still unknown. We have therefore focused our attention on determination of the influence of acute immobilization stress (IMO) in normal mice and mice deficient in corticotropin-releasing hormone (CRH). Specifically, we have analyzed levels of hyperphosphorylated tau proteins, bearing the AD-specific phospho-epitopes (AT-8, pT181, and PHF-1), which are implicated in the pathogenesis of AD. We found that IMO induces transient hyperphosphorylation of tau proteins regardless of continuation of the stimulus. Concerning tau modifications, detailed analysis of the mouse brain revealed that neurons in different brain regions including frontal cortex, temporal cortex, hippocampal C1 and CA3 regions, dentate gyrus as well as nucleus basalis Meynert, and several brainstem nuclei such as locus coeruleus but also raphe nucleus and substantia nigra respond similarly to IMO. The strongest tau protein phosphorylation was observed after 30 min of IMO stress. Stress lasting for 120 min led either to the disappearance of tau hyperphosphorylation or to the induction of a second wave of hyperphosphorylation. Noteworthy is the magnitude of pathological phosphorylation of tau protein in CRH and glucocorticoids deficient mice, being much lower in comparison to that observed in wild-type animals, which suggests a critical role of CRH in the pathogenesis of AD. Thus, our results indicate that hyperphosphorylation of tau protein induced by stress may represent the pathogenic event upstream of tau protein misfolding, which leads to progression or eventually initiation of neurodegeneration. The data show that CRH plays an important role in stress induced hyperphosphorylation of tau protein, which might be either a direct effect of CRH innervations in the brain or an effect mediated via the hypothalamo-pituitary-adrenal axis.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Hormônio Liberador da Corticotropina/deficiência , Estresse Fisiológico , Regulação para Cima , Proteínas tau/metabolismo , Doença de Alzheimer/imunologia , Animais , Biomarcadores/metabolismo , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Hormônio Liberador da Corticotropina/metabolismo , Epitopos/imunologia , Imobilização , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Proteínas tau/imunologia
14.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 10): 1181-5, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23027743

RESUMO

The Alzheimer's disease-associated protein tau is an intrinsically disordered protein with no preferred structure in solution. Under physiological conditions, tau binds to microtubules and regulates their dynamics, whereas during the development of neurodegeneration tau dissociates from microtubules, misfolds and creates highly insoluble deposits. To elucidate the determinants of tau-protein misfolding, tau peptides from microtubule-binding motifs were crystallized in complexes with Fab fragments of specific monoclonal antibodies. The crystals diffracted to 1.69 Šresolution and gave complete data sets using a synchrotron X-ray source. Molecular replacement was used to solve the phase problem.


Assuntos
Fragmentos Fab das Imunoglobulinas/química , Proteínas tau/química , Motivos de Aminoácidos , Anticorpos Monoclonais , Cristalização , Cristalografia por Raios X , Fragmentos Fab das Imunoglobulinas/imunologia , Microtúbulos/química , Microtúbulos/metabolismo , Estrutura Terciária de Proteína , Proteínas tau/imunologia , Proteínas tau/metabolismo
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 10): 1186-90, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23027744

RESUMO

The major constituent of the Alzheimer's disease paired helical filaments (PHF) core is the intrinsically disordered protein (IDP) tau. Globular binding partners, e.g. monoclonal antibodies, can stabilize the fold of disordered tau in complexes. A previously published structure of a proteolytically generated tau fragment in a complex with the PHF-specific monoclonal antibody MN423 revealed a turn-like structure of the PHF core C-terminus [Sevcik et al. (2007). FEBS Lett. 581, 5872-5878]. To examine the structures of longer better-defined PHF segments, crystals of the MN423 Fab fragment were grown in the presence of two synthetic peptides derived from the PHF core C-terminus. For each, X-ray diffraction data were collected at 100 K at a synchrotron source and initial phases were obtained by molecular replacement.


Assuntos
Doença de Alzheimer , Anticorpos Monoclonais/química , Fragmentos Fab das Imunoglobulinas/química , Fragmentos de Peptídeos/química , Doença de Alzheimer/imunologia , Animais , Anticorpos Monoclonais/imunologia , Cristalização , Cristalografia por Raios X , Epitopos/química , Epitopos/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Camundongos , Fragmentos de Peptídeos/imunologia , Estrutura Secundária de Proteína , Proteínas tau/química
16.
Neurodegener Dis ; 10(1-4): 242-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22433908

RESUMO

BACKGROUND: Neurodegeneration induced by misfolded tau protein and neuroinflammation represent the major hallmarks of human tauopathies including Alzheimer's disease (AD). While tau driven neurodegeneration significantly correlates with disease progression, inflammation is considered to be an important factor regulating the resistance or susceptibility to AD. The emerging evidence suggests that the genes related to immunity can influence neurodegeneration. OBJECTIVE: In order to determine the role of MHC class II in the tau neurofibrillary cascade, we generated and used transgenic lines expressing human truncated tau protein in either spontaneously hypertensive rat (SHR) or Wistar-Kyoto rat (WKY) genetic background. METHODS: Brains of WKY and SHR transgenic rats and their age-matched nontransgenic littermates were examined by immunohistochemistry and RT-PCR. RESULTS: Our results clearly showed that genetic background determined the inflammatory pattern (MHC class II) induced by tau neurodegenerative cascade in the transgenic rats expressing human misfolded truncated tau. CONCLUSION: Using two transgenic rat lines with different immunogenetic backgrounds, expressing the same transgene, we conclude that genetic background is a potent modifier of the type of the immune response induced by tau neurodegeneration.


Assuntos
Doença de Alzheimer/complicações , Antígenos de Histocompatibilidade Classe II/metabolismo , Transtornos da Memória/etiologia , Emaranhados Neurofibrilares/patologia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Transtornos da Memória/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Transgênicos
17.
EBioMedicine ; 76: 103818, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35078012

RESUMO

BACKGROUND: The emergence of new SARS-CoV-2 variants of concern B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma) and B.1.617.2 (Delta) that harbor mutations in the viral S protein raised concern about activity of current vaccines and therapeutic antibodies. Independent studies have shown that mutant variants are partially or completely resistant against some of the therapeutic antibodies authorized for emergency use. METHODS: We employed hybridoma technology, ELISA-based and cell-based S-ACE2 interaction assays combined with authentic virus neutralization assays to develop second-generation antibodies, which were specifically selected for their ability to neutralize the new variants of SARS-CoV-2. FINDINGS: AX290 and AX677, two monoclonal antibodies with non-overlapping epitopes, exhibit subnanomolar or nanomolar affinities to the receptor binding domain of the viral Spike protein carrying amino acid substitutions N501Y, N439K, E484K, K417N, and a combination N501Y/E484K/K417N found in the circulating virus variants. The antibodies showed excellent neutralization of an authentic SARS-CoV-2 virus representing strains circulating in Europe in spring 2020 and also the variants of concern B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.2 (Delta). In addition, AX677 is able to bind Omicron Spike protein just like the wild type Spike. The combination of the two antibodies prevented the appearance of escape mutations of the authentic SARS-CoV-2 virus. Prophylactic administration of AX290 and AX677, either individually or in combination, effectively reduced viral burden and inflammation in the lungs, and prevented disease in a mouse model of SARS-CoV-2 infection. INTERPRETATION: The virus-neutralizing properties were fully reproduced in chimeric mouse-human versions of the antibodies, which may represent a promising tool for COVID-19 therapy. FUNDING: The study was funded by AXON Neuroscience SE and AXON COVIDAX a.s.


Assuntos
Anticorpos Monoclonais/imunologia , Antineoplásicos Imunológicos/imunologia , Epitopos Imunodominantes/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Deriva e Deslocamento Antigênicos , Antineoplásicos Imunológicos/uso terapêutico , COVID-19/virologia , Modelos Animais de Doenças , Humanos , Cinética , Pulmão/patologia , Camundongos , Mutação , Testes de Neutralização , Ligação Proteica , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Tratamento Farmacológico da COVID-19
18.
Nat Aging ; 1(6): 521-534, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-37117834

RESUMO

Alzheimer's disease (AD) pathology is partly characterized by accumulation of aberrant forms of tau protein. Here we report the results of ADAMANT, a 24-month double-blinded, parallel-arm, randomized phase 2 multicenter placebo-controlled trial of AADvac1, an active peptide vaccine designed to target pathological tau in AD (EudraCT 2015-000630-30). Eleven doses of AADvac1 were administered to patients with mild AD dementia at 40 µg per dose over the course of the trial. The primary objective was to evaluate the safety and tolerability of long-term AADvac1 treatment. The secondary objectives were to evaluate immunogenicity and efficacy of AADvac1 treatment in slowing cognitive and functional decline. A total of 196 patients were randomized 3:2 between AADvac1 and placebo. AADvac1 was safe and well tolerated (AADvac1 n = 117, placebo n = 79; serious adverse events observed in 17.1% of AADvac1-treated individuals and 24.1% of placebo-treated individuals; adverse events observed in 84.6% of AADvac1-treated individuals and 81.0% of placebo-treated individuals). The vaccine induced high levels of IgG antibodies. No significant effects were found in cognitive and functional tests on the whole study sample (Clinical Dementia Rating-Sum of the Boxes scale adjusted mean point difference -0.360 (95% CI -1.306, 0.589)), custom cognitive battery adjusted mean z-score difference of 0.0008 (95% CI -0.169, 0.172). We also present results from exploratory and post hoc analyses looking at relevant biomarkers and clinical outcomes in specific subgroups. Our results show that AADvac1 is safe and immunogenic, but larger stratified studies are needed to better evaluate its potential clinical efficacy and impact on disease biomarkers.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/terapia , Proteínas tau , Imunoterapia Ativa/métodos , Biomarcadores
19.
J Struct Biol ; 171(1): 74-81, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20184958

RESUMO

Flexibility of intrinsically disordered tau protein is important for performing its functions. It is believed that alteration of the flexibility is instrumental to the assembly of tau protein into paired helical filaments (PHF) in tauopathies. Tau flexibility represents the main obstacle for structure determination of its conformation in physiology and/or pathology. We have alleviated this inherited difficulty by using specific monoclonal antibodies as tau protein surrogate binding partners. In this work we compare two "antibody mold structures": (1) X-ray structure of the free form of the Alzheimer's disease PHF core-specific antibody MN423 and (2) previously solved structure of the complex of MN423 with the PHF core C-terminal tau peptide. We found that MN423 combining site is in both structures identical. As a consequence, recombinant tau assumes in the complex a fold determined by the antibody combining site. Obtained results show that MN423 functions as a molecular mold for the PHF core segment, and opens the way for structure determination of other PHF core segments providing that other conformation-specific antibodies are available. Data from in silico docking of tau peptide into antibody mold, obtained in this study, show that biochemical data and computational approaches provide results comparable to X-ray crystallography.


Assuntos
Anticorpos Monoclonais/química , Proteínas tau/química , Cristalografia por Raios X , Modelos Moleculares , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química
20.
J Neuroinflammation ; 7: 64, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20937161

RESUMO

BACKGROUND: Numerous epidemiological studies demonstrate that genetic background modifies the onset and the progression of Alzheimer's disease and related neurodegenerative disorders. The efficacious influence of genetic background on the disease pathway of amyloid beta has been meticulously described in rodent models. Since the impact of genetic modifiers on the neurodegenerative and neuroinflammatory cascade induced by misfolded tau protein is yet to be elucidated, we have addressed the issue by using transgenic lines expressing the same human truncated tau protein in either spontaneously hypertensive rat (SHR) or Wistar-Kyoto (WKY) genetic background. METHODS: Brains of WKY and SHR transgenic rats in the terminal stage of phenotype and their age-matched non-transgenic littermates were examined by means of immunohistochemistry and unbiased stereology. Basic measures of tau-induced neurodegeneration (load of neurofibrillary tangles) and neuroinflammation (number of Iba1-positive microglia, their activated morphology, and numbers of microglia immunoreactive for MHCII and astrocytes immunoreactive for GFAP) were quantified with an optical fractionator in brain areas affected by neurofibrillary pathology (pons, medulla oblongata). The stereological data were evaluated using two-way ANOVA and Student's t-test. RESULTS: Tau neurodegeneration (neurofibrillary tangles (NFTs), axonopathy) and neuroinflammation (microgliosis, astrocytosis) appeared in both WKY and SHR transgenic rats. Although identical levels of transgene expression in both lines were present, terminally-staged WKY transgenic rats displayed significantly lower final NFT loads than their SHR transgenic counterparts. Interestingly, microglial responses showed a striking difference between transgenic lines. Only 1.6% of microglia in SHR transgenic rats expressed MHCII in spite of having a robust phagocytic phenotype, whereas in WKY transgenic rats, 23.2% of microglia expressed MHCII despite displaying a considerably lower extent of transformation into phagocytic phenotype. CONCLUSIONS: These results show that the immune response represents a pivotal and genetically variable modifying factor that is able to influence vulnerability to neurodegeneration. Therefore, targeted immunomodulation could represent a prospective therapeutic approach to Alzheimer's disease.


Assuntos
Encéfalo/metabolismo , Encefalite/genética , Degeneração Neural/genética , Tauopatias/genética , Proteínas tau/metabolismo , Análise de Variância , Animais , Western Blotting , Encéfalo/patologia , Contagem de Células , Encefalite/metabolismo , Encefalite/patologia , Imuno-Histoquímica , Masculino , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Transgênicos , Coloração pela Prata , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA