Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Neurodegener ; 16(1): 39, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172073

RESUMO

BACKGROUND: For decades, dementia has been characterized by accumulation of waste in the brain and low-grade inflammation. Over the years, emerging studies highlighted the involvement of the immune system in neurodegenerative disease emergence and severity. Numerous studies in animal models of amyloidosis demonstrated the beneficial role of monocyte-derived macrophages in mitigating the disease, though less is known regarding tauopathy. Boosting the immune system in animal models of both amyloidosis and tauopathy, resulted in improved cognitive performance and in a reduction of pathological manifestations. However, a full understanding of the chain of events that is involved, starting from the activation of the immune system, and leading to disease mitigation, remained elusive. Here, we hypothesized that the brain-immune communication pathway that is needed to be activated to combat tauopathy involves monocyte mobilization via the C-C chemokine receptor 2 (CCR2)/CCL2 axis, and additional immune cells, such as CD4+ T cells, including FOXP3+ regulatory CD4+ T cells. METHODS: We used DM-hTAU transgenic mice, a mouse model of tauopathy, and applied an approach that boosts the immune system, via blocking the inhibitory Programmed cell death protein-1 (PD-1)/PD-L1 pathway, a manipulation previously shown to alleviate disease symptoms and pathology. An anti-CCR2 monoclonal antibody (αCCR2), was used to block the CCR2 axis in a protocol that partially eliminates monocytes from the circulation at the time of anti-PD-L1 antibody (αPD-L1) injection, and for the critical period of their recruitment into the brain following treatment. RESULTS: Performance of DM-hTAU mice in short-term and working memory tasks, revealed that the beneficial effect of αPD-L1, assessed 1 month after a single injection, was abrogated following blockade of CCR2. This was accompanied by the loss of the beneficial effect on disease pathology, assessed by measurement of cortical aggregated human tau load using Homogeneous Time Resolved Fluorescence-based immunoassay, and by evaluation of hippocampal neuronal survival. Using both multiparametric flow cytometry, and Cytometry by Time Of Flight, we further demonstrated the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, 12 days following the treatment, which was absent subsequent to CCR2 blockade. In addition, measurement of hippocampal levels of the T-cell chemoattractant, C-X-C motif chemokine ligand 12 (Cxcl12), and of inflammatory cytokines, revealed that αPD-L1 treatment reduced their expression, while blocking CCR2 reversed this effect. CONCLUSIONS: The CCR2/CCL2 axis is required to modify pathology using PD-L1 blockade in a mouse model of tauopathy. This modification involves, in addition to monocytes, the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, and the T-cell chemoattractant, Cxcl12.


Assuntos
Quimiocina CCL2/metabolismo , Receptores CCR2/metabolismo , Tauopatias/imunologia , Tauopatias/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Quimiocina CCL2/imunologia , Modelos Animais de Doenças , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Receptores CCR2/imunologia , Tauopatias/patologia
2.
J Alzheimers Dis ; 79(2): 683-691, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33337376

RESUMO

BACKGROUND: Anti-IgLON5 disease is a rare neurodegenerative tauopathy that displays heterogeneity in clinical spectrum, disease course, cerebrospinal fluid (CSF) findings, and variable response to immunotherapy. Sleep disorders, bulbar dysfunction, and gait abnormalities are common presenting symptoms, and conventional brain MRI scanning is often unrevealing. OBJECTIVE: To provide a comprehensive overview of the literature and to assess the frequency of symptoms, MRI findings, and treatment response in patients with IgLON5 autoimmunity in the serum and CSF or restricted to serum. METHODS: We examined a 65-year-old woman with bulbar-onset IgLON5 disease with serum-restricted antibodies, and we also performed a systematic review of all confirmed cases reported in the English literature. RESULTS: We identified 93 patients, included our case. Clinical data were obtained in 58 subjects, in whom the most frequent symptoms were sleep-disordered breathing, dysphagia, parasomnias, dysarthria, limb or gait ataxia, stridor or vocal cord paresis, movement disorders, and postural instability. Distinct MRI alterations were identified in 12.5% of cases, as opposed to unspecific or unremarkable changes in the remaining patients. T2-hyperintense non-enhancing signal alterations involving the hypothalamus and the brainstem tegmentum were observed only in the present case. Inflammatory CSF was found in half of the cases and serum-restricted antibodies in 4 patients. Treatment with immunosuppressant or immunomodulatory drugs led to sustained clinical response in 19/52 patients. CONCLUSION: Anti-IgLON5 autoimmunity should be considered in patients with sleep disorders, bulbar syndrome, autonomic involvement, and movement disorders, and high-field brain MRI can be of diagnostic help.


Assuntos
Doenças Autoimunes/imunologia , Paralisia Bulbar Progressiva/imunologia , Moléculas de Adesão Celular Neuronais/imunologia , Doenças Hipotalâmicas/imunologia , Doenças Neurodegenerativas/imunologia , Tauopatias/imunologia , Idoso , Doenças Autoimunes/diagnóstico por imagem , Doenças Autoimunes/patologia , Paralisia Bulbar Progressiva/diagnóstico por imagem , Paralisia Bulbar Progressiva/patologia , Feminino , Humanos , Doenças Hipotalâmicas/diagnóstico por imagem , Doenças Hipotalâmicas/patologia , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/patologia , Tauopatias/diagnóstico por imagem , Tauopatias/patologia
3.
Front Immunol ; 11: 997, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508844

RESUMO

Innate immune activation is a major contributor to Alzheimer's Disease (AD) pathophysiology, although the mechanisms involved are poorly understood. Chemokine C-C motif ligand (CCL) 2 is produced by neurons and glial cells and is upregulated in the AD brain. Transgene expression of CCL2 in mouse models of amyloidosis produces microglia-induced amyloid ß oligomerization, a strong indication of the role of these activation pathways in the amyloidogenic processes of AD. We have previously shown that CCL2 polarizes microglia in wild type mice. However, how CCL2 signaling contributes to tau pathogenesis remains unknown. To address this question, CCL2 was delivered via recombinant adeno-associated virus serotype 9 into both cortex and hippocampus of a mouse model with tau pathology (rTg4510). We report that CCL2 overexpression aggravated tau pathology in rTg4510 as shown by the increase in Gallyas stained neurofibrillary tangles as well as phosphorylated tau-positive inclusions. In addition, biochemical analysis showed a reduction in the levels of detergent-soluble tau species followed by increase in the insoluble fraction, indicating a shift toward larger tau aggregates. Indeed, increased levels of high molecular weight species of phosphorylated tau were found in the mice injected with CCL2. We also report that worsening of tau pathology following CCL2 overexpression was accompanied by a distinct inflammatory response. We report an increase in leukocyte common antigen (CD45) and Cluster of differentiation 68 (CD68) expression in the brain of rTg4510 mice without altering the expression levels of a cell-surface protein Transmembrane Protein 119 (Tmem119) and ionized calcium-binding adaptor molecule 1 (Iba-1) in resident microglia. Furthermore, the analysis of cytokines in brain extract showed a significant increase in interleukin (IL)-6 and CCL3, while CCL5 levels were decreased in CCL2 mice. No changes were observed in IL-1α, IL-1ß, TNF-α. IL-4, Vascular endothelial growth factor-VEGF, IL-13 and CCL11. Taken together our data report for the first time that overexpression of CCL2 promotes the increase of pathogenic tau species and is associated with glial neuroinflammatory changes that are deleterious. We propose that these events may contribute to the pathogenesis of Alzheimer's disease and other tauopathies.


Assuntos
Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Neuroglia/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/patologia , Quimiocina CCL2/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Transgênicos , Mutação , Neuroglia/imunologia , Neuroglia/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Transdução de Sinais , Tauopatias/genética , Tauopatias/imunologia , Tauopatias/patologia , Regulação para Cima , Proteínas tau/genética
4.
Cell Rep ; 28(8): 2111-2123.e6, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31433986

RESUMO

Complement pathway overactivation can lead to neuronal damage in various neurological diseases. Although Alzheimer's disease (AD) is characterized by ß-amyloid plaques and tau tangles, previous work examining complement has largely focused on amyloidosis models. We find that glial cells show increased expression of classical complement components and the central component C3 in mouse models of amyloidosis (PS2APP) and more extensively tauopathy (TauP301S). Blocking complement function by deleting C3 rescues plaque-associated synapse loss in PS2APP mice and ameliorates neuron loss and brain atrophy in TauP301S mice, improving neurophysiological and behavioral measurements. In addition, C3 protein is elevated in AD patient brains, including at synapses, and levels and processing of C3 are increased in AD patient CSF and correlate with tau. These results demonstrate that complement activation contributes to neurodegeneration caused by tau pathology and suggest that blocking C3 function might be protective in AD and other tauopathies.


Assuntos
Doença de Alzheimer/imunologia , Amiloidose/imunologia , Complemento C3/metabolismo , Degeneração Neural/imunologia , Tauopatias/imunologia , Doença de Alzheimer/genética , Animais , Atrofia , Comportamento Animal , Biomarcadores/metabolismo , Encéfalo/patologia , Complemento C1q/metabolismo , Complemento C3/líquido cefalorraquidiano , Complemento C3/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Transgênicos , Degeneração Neural/genética , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/metabolismo , Sinapses/metabolismo
5.
Acta Neuropathol Commun ; 6(1): 82, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30134961

RESUMO

Tau, the main component of the neurofibrillary tangles (NFTs), is an attractive target for immunotherapy in Alzheimer's disease (AD) and other tauopathies. MC1/Alz50 are currently the only antibodies targeting a disease-specific conformational modification of tau. Passive immunization experiments using intra-peritoneal injections have previously shown that MC1 is effective at reducing tau pathology in the forebrain of tau transgenic JNPL3 mice. In order to reach a long-term and sustained brain delivery, and avoid multiple injection protocols, we tested the efficacy of the single-chain variable fragment of MC1 (scFv-MC1) to reduce tau pathology in the same animal model, with focus on brain regional differences. ScFv-MC1 was cloned into an AAV delivery system and was directly injected into the hippocampus of adult JNPL3 mice. Specific promoters were employed to selectively target neurons or astrocytes for scFv-MC1 expression. ScFv-MC1 was able to decrease soluble, oligomeric and insoluble tau species, in our model. The effect was evident in the cortex, hippocampus and hindbrain. The astrocytic machinery appeared more efficient than the neuronal, with significant reduction of pathology in areas distant from the site of injection. To our knowledge, this is the first evidence that an anti-tau conformational scFv antibody, delivered directly into the mouse adult brain, is able to reduce pathological tau, providing further insight into the nature of immunotherapy strategies.


Assuntos
Anticorpos/farmacologia , Encéfalo/efeitos dos fármacos , Tauopatias/imunologia , Tauopatias/terapia , Proteínas tau/imunologia , Proteínas tau/metabolismo , Análise de Variância , Animais , Anticorpos/sangue , Complexo Antígeno-Anticorpo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Dependovirus , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Parvovirinae/genética , Conformação Proteica/efeitos dos fármacos , Tauopatias/genética , Proteínas tau/genética
6.
J Exp Med ; 214(5): 1227-1238, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28416651

RESUMO

Tauopathies are characterized by the progressive accumulation of hyperphosphorylated, aggregated forms of tau. Our laboratory has previously demonstrated that passive immunization with an anti-tau antibody, HJ8.5, decreased accumulation of pathological tau in a human P301S tau-expressing transgenic (P301S-tg) mouse model of frontotemporal dementia/tauopathy. To investigate whether the Fc domain of HJ8.5 is required for the therapeutic effect, we engineered single-chain variable fragments (scFvs) derived from HJ8.5 with variable linker lengths, all specific to human tau. Based on different binding properties, we selected two anti-tau scFvs and tested their efficacy in vivo by adeno-associated virus-mediated gene transfer to the brain of P301S-tg mice. The scFvs significantly reduced levels of hyperphosphorylated, aggregated tau in brain tissue of P301S-tg mice, associated with a decrease in detergent-soluble tau species. Interestingly, these mice showed substantial levels of scFvs in the cerebrospinal fluid without significant effects on total extracellular tau levels. Therefore, our study provides a novel strategy for anti-tau immunotherapeutics that potentially limits a detrimental proinflammatory response.


Assuntos
Anticorpos de Cadeia Única/imunologia , Tauopatias/imunologia , Proteínas tau/imunologia , Animais , Encéfalo/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/fisiologia , Tauopatias/metabolismo
7.
J Neurosci ; 36(49): 12425-12435, 2016 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-27927959

RESUMO

Passive immunization with anti-tau monoclonal antibodies has been shown by several laboratories to reduce age-dependent tau pathology and neurodegeneration in mutant tau transgenic mice. These studies have used repeated high weekly doses of various tau antibodies administered systemically for several months and have reported reduced tau pathology of ∼40-50% in various brain regions. Here we show that direct intrahippocampal administration of the adeno-associated virus (AAV)-vectored anti-phospho-tau antibody PHF1 to P301S tau transgenic mice results in high and durable antibody expression, primarily in neurons. Hippocampal antibody levels achieved after AAV delivery were ∼50-fold more than those reported following repeated systemic administration. In contrast to systemic passive immunization, we observed markedly reduced (≥80-90%) hippocampal insoluble pathological tau species and neurofibrillary tangles following a single dose of AAV-vectored PHF1 compared with mice treated with an AAV-IgG control vector. Moreover, the hippocampal atrophy observed in untreated P301S mice was fully rescued by treatment with the AAV-vectored PHF1 antibody. Vectored passive immunotherapy with an anti-tau monoclonal antibody may represent a viable therapeutic strategy for treating or preventing such tauopathies as frontotemporal dementia, progressive supranuclear palsy, or Alzheimer's disease. SIGNIFICANCE STATEMENT: We have used an adeno-associated viral (AAV) vector to deliver the genes encoding an anti-phospho-tau monoclonal antibody, PHF1, directly to the brain of mice that develop neurodegeneration due to a tau mutation that causes frontotemporal dementia (FTD). When administered systemically, PHF1 has been shown to modestly reduce tau pathology and neurodegeneration. Since such antibodies do not readily cross the blood-brain barrier, we used an AAV vector to deliver antibody directly to the hippocampus and observed much higher antibody levels and a much greater reduction in tau pathology. Using AAV vectors to deliver antibodies like PHF1 directly to brain may constitute a novel approach to treating various neurodegenerative disorders, such as FTD and Alzheimer's disease.


Assuntos
Anticorpos Monoclonais/imunologia , Imunização Passiva/métodos , Tauopatias/imunologia , Tauopatias/prevenção & controle , Fatores de Transcrição/imunologia , Proteínas tau/genética , Animais , Anticorpos Monoclonais/administração & dosagem , Proteínas de Ligação a DNA , Dependovirus/imunologia , Feminino , Hipocampo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microinjeções , Mutação/genética , Emaranhados Neurofibrilares/patologia , Proteínas do Grupo Polycomb , Distribuição Tecidual
8.
Curr Opin Neurol ; 29(4): 496-506, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27262149

RESUMO

PURPOSE OF REVIEW: This review highlights the recent developments in immune-mediated movement disorders and how they reflect on clinical practice and our understanding of the underlying pathophysiological mechanisms. RECENT FINDINGS: The antibody spectrum associated with stiff person syndrome and related disorders (SPSD) has broadened and, apart from the classic glutamic acid decarboxylase (GAD)- and amphiphysin-antibodies, includes now also antibodies against dipeptidyl-peptidase-like protein-6 (DPPX), gamma-aminobutyric acid type A receptor (GABAAR), glycine receptor (GlyR) and glycine transporter 2 (GlyT2). The field of movement disorders with neuronal antibodies keeps expanding with the discovery for example of antibodies against leucine rich glioma inactivated protein 1 (LGI1) and contactin associated protein 2 (Caspr2) in chorea, or antibodies targeting ARHGAP26- or Na/K ATPase alpha 3 subunit (ATP1A3) in cerebellar ataxia. Moreover, neuronal antibodies may partly account for movement disorders attributed for example to Sydenham's chorea, coeliac disease, or steroid responsive encephalopathy with thyroid antibodies. Lastly, there is an interface of immunology, genetics and neurodegeneration, e.g. in Aicardi-Goutières syndrome or the tauopathy with IgLON5-antibodies. SUMMARY: Clinicians should be aware of new antibodies such as dipeptidyl-peptidase-like protein-6, gamma-aminobutyric acid type A receptor and glycine transporter 2 in stiff person syndrome and related disorders, as well as of the expanding spectrum of immune-mediated movement disorders.


Assuntos
Rigidez Muscular Espasmódica/imunologia , Tauopatias/imunologia , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/imunologia , Autoimunidade/genética , Humanos , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/imunologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/imunologia , Neurônios/imunologia , Rigidez Muscular Espasmódica/fisiopatologia
9.
J Biol Chem ; 289(29): 19855-61, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24860099

RESUMO

Work over the past 4 years indicates that multiple proteins associated with neurodegenerative diseases, especially Tau and α-synuclein, can propagate aggregates between cells in a prion-like manner. This means that once an aggregate is formed it can escape the cell of origin, contact a connected cell, enter the cell, and induce further aggregation via templated conformational change. The prion model predicts a key role for extracellular protein aggregates in mediating progression of disease. This suggests new therapeutic approaches based on blocking neuronal uptake of protein aggregates and promoting their clearance. This will likely include therapeutic antibodies or small molecules, both of which can be developed and optimized in vitro prior to preclinical studies.


Assuntos
Príons/metabolismo , Tauopatias/metabolismo , Tauopatias/terapia , Proteínas tau/metabolismo , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Membrana Celular/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Príons/química , Conformação Proteica , Multimerização Proteica , Transporte Proteico , Tauopatias/imunologia , Vacinas/imunologia , Vacinas/uso terapêutico , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Proteínas tau/química , Proteínas tau/imunologia
10.
Expert Rev Vaccines ; 13(3): 429-41, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24405291

RESUMO

Plants are considered advantageous platforms for biomanufacturing recombinant vaccines. This constitutes a field of intensive research and some plant-derived vaccines are expected to be marketed in the near future. In particular, plant-based production of immunogens targeting molecules with implications on the pathology of Alzheimer's has been explored over the last decade. These efforts involve targeting amyloid beta and ß-secretase with several immunogen configurations that have been evaluated in test animals. The results of these developments are analyzed in this review. Perspectives on the topic are identified, such as exploring additional antigen configurations and adjuvants in order to improve immunization schemes, characterizing in detail the elicited immune responses, and immunological considerations in the achievement of therapeutic humoral responses via mucosal immunization. Safety concerns related to these therapies will also be discussed.


Assuntos
Doença de Alzheimer/prevenção & controle , Vacinas contra Alzheimer/imunologia , Secretases da Proteína Precursora do Amiloide/imunologia , Peptídeos beta-Amiloides/imunologia , Fitoterapia/métodos , Doença de Alzheimer/imunologia , Animais , Humanos , Imunoterapia/métodos , Solanum lycopersicum/imunologia , Solanum lycopersicum/metabolismo , Camundongos , Oryza/imunologia , Oryza/metabolismo , Vírus de Plantas/genética , Solanum tuberosum/imunologia , Solanum tuberosum/metabolismo , Tauopatias/genética , Tauopatias/imunologia , Nicotiana/imunologia , Nicotiana/metabolismo , Vacinação , Proteínas tau/genética , Proteínas tau/imunologia
11.
PLoS One ; 8(8): e72301, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977276

RESUMO

Progressive aggregation of protein Tau into oligomers and fibrils correlates with cognitive decline and synaptic dysfunction, leading to neurodegeneration in vulnerable brain regions in Alzheimer's disease. The unmet need of effective therapy for Alzheimer's disease, combined with problematic pharmacological approaches, led the field to explore immunotherapy, first against amyloid peptides and recently against protein Tau. Here we adapted the liposome-based amyloid vaccine that proved safe and efficacious, and incorporated a synthetic phosphorylated peptide to mimic the important phospho-epitope of protein Tau at residues pS396/pS404. We demonstrate that the liposome-based vaccine elicited, rapidly and robustly, specific antisera in wild-type mice and in Tau.P301L mice. Long-term vaccination proved to be safe, because it improved the clinical condition and reduced indices of tauopathy in the brain of the Tau.P301L mice, while no signs of neuro-inflammation or other adverse neurological effects were observed. The data corroborate the hypothesis that liposomes carrying phosphorylated peptides of protein Tau have considerable potential as safe and effective treatment against tauopathies, including Alzheimer's disease.


Assuntos
Vacinas contra Alzheimer/imunologia , Anticorpos Neutralizantes/sangue , Peptídeos/imunologia , Fosfoproteínas/imunologia , Tauopatias/tratamento farmacológico , Proteínas tau/imunologia , Vacinas contra Alzheimer/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Humanos , Lipossomos/química , Camundongos , Camundongos Transgênicos , Peptídeos/administração & dosagem , Peptídeos/síntese química , Fosfoproteínas/administração & dosagem , Fosfoproteínas/síntese química , Fosforilação , Desempenho Psicomotor/efeitos dos fármacos , Tauopatias/imunologia , Tauopatias/fisiopatologia , Resultado do Tratamento , Vacinação , Proteínas tau/antagonistas & inibidores , Proteínas tau/genética
12.
Neuropathol Appl Neurobiol ; 29(3): 288-302, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787326

RESUMO

Pathological inclusions containing fibrillar aggregates of hyperphosphorylated tau protein are a characteristic feature in the tauopathies, which include Alzheimer's disease, frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), progressive supranuclear palsy, corticobasal degeneration and Pick's disease. Tau isoform composition and cellular and regional distribution as well as morphology of these inclusions vary in each disorder. Recently, several pathological missense and exon 10 splice-donor site mutations of the tau gene were identified in FTDP-17. Exon 10 codes for the second of four microtubule-binding repeat domains. The splice-site mutations result in increased inclusion of exon 10 which causes a relative increase in tau isoforms containing four microtubule-binding repeat domains over those containing three repeat domains. This could be a central aetiological mechanism in FTDP-17 and, perhaps, other related tauopathies. We have investigated changes in the ratio and distribution of three-repeat and four-repeat tau in the different tauopathies as a basis of the phenotypic range of these disorders and the selective vulnerability of different subsets of neurones. In this study, we have developed two monoclonal antibodies, RD3 and RD4 that effectively distinguish these closely related tau isoforms. These new isoform-specific antibodies are useful tools for analysing tau isoform expression and distribution as well as pathological changes in the human brain.


Assuntos
Anticorpos Monoclonais , Especificidade de Anticorpos , Encéfalo/patologia , Tauopatias/imunologia , Proteínas tau/análise , Proteínas tau/imunologia , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Linhagem Celular , Humanos , Imuno-Histoquímica , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Corpos de Inclusão/ultraestrutura , Microscopia Imunoeletrônica , Microtúbulos/química , Neuroblastoma/química , Neuroblastoma/genética , Emaranhados Neurofibrilares/química , Neurônios/metabolismo , Neurônios/patologia , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Ratos , Proteínas Recombinantes , Tauopatias/genética , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/biossíntese , Proteínas tau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA