Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Aging Neurosci ; 14: 974414, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36062141

RESUMO

The conversion and transmission of misfolded proteins established the basis for the prion concept. Neurodegenerative diseases are considered "prion-like" disorders that lack infectivity. Among them, tauopathies are characterized by the conversion of native tau protein into an abnormally folded aggregate. During the progression of the disease, misfolded tau polymerizes into oligomers and intracellular neurofibrillary tangles (NFTs). While the toxicity of NFTs is an ongoing debate, the contribution of tau oligomers to early onset neurodegenerative pathogenesis is accepted. Tau oligomers are readily transferred from neuron to neuron propagating through the brain inducing neurodegeneration. Recently, transmission of tau oligomers via exosomes is now proposed. There is still too much to uncover about tau misfolding and propagation. Here we summarize novel findings of tau oligomers transmission and propagation via exosomes.

2.
Brain Commun ; 2(1): fcaa016, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32954281

RESUMO

This scientific commentary refers to 'Analysis of α-synuclein species enriched from cerebral cortex of humans with sporadic dementia with Lewy bodies', by Sanderson et al. (https://doi.org/10.1093/braincomms/fcaa010).

3.
Front Immunol ; 9: 2703, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30515174

RESUMO

Ataxia-telangiectasia (A-T) is a complex disease arising from mutations in the ATM gene (Ataxia-Telangiectasia Mutated), which plays crucial roles in repairing double-strand DNA breaks (DSBs). Heterogeneous immunodeficiency, extreme radiosensitivity, frequent appearance of tumors and neurological degeneration are hallmarks of the disease, which carries high morbidity and mortality because only palliative treatments are currently available. Gene therapy was effective in animal models of the disease, but the large size of the ATM cDNA required the use of HSV-1 or HSV/AAV hybrid amplicon vectors, whose characteristics make them unlikely tools for treating A-T patients. Due to recent advances in vector packaging, production and biosafety, we developed a lentiviral vector containing the ATM cDNA and tested whether or not it could rescue cellular defects of A-T human mutant fibroblasts. Although the cargo capacity of lentiviral vectors is an inherent limitation in their use, and despite the large size of the transgene, we successfully transduced around 20% of ATM-mutant cells. ATM expression and phosphorylation assays indicated that the neoprotein was functional in transduced cells, further reinforced by their restored capacity to phosphorylate direct ATM substrates such as p53 and their capability to repair radiation-induced DSBs. In addition, transduced cells also restored cellular radiosensitivity and cell cycle abnormalities. Our results demonstrate that lentiviral vectors can be used to rescue the intrinsic cellular defects of ATM-mutant cells, which represent, in spite of their limitations, a proof-of-concept for A-T gene therapy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia , Ataxia Telangiectasia , Fibroblastos , Vetores Genéticos , Lentivirus , Mutação , Transdução Genética , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Ataxia Telangiectasia/patologia , Proteínas Mutadas de Ataxia Telangiectasia/biossíntese , Proteínas Mutadas de Ataxia Telangiectasia/genética , Linhagem Celular , Fibroblastos/metabolismo , Fibroblastos/patologia
4.
Mol Neurodegener ; 13(1): 13, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29544548

RESUMO

BACKGROUND: We have evaluated the efficacy of targeting the toxic, oligomeric form of tau protein by passive immunotherapy in a mouse model of synucleinopathy. Parkinson's disease and Lewy body dementia are two of the most common neurodegenerative disorders and are primarily characterized by the accumulation of α-synuclein in Lewy bodies. However, evidence shows that smaller, oligomeric aggregates are likely the most toxic form of the protein. Moreover, a large body of research suggests that α-synuclein interacts with tau in disease and may act in a synergistic mechanism, implicating tau oligomers as a potential therapeutic target. METHODS: We treated seven-month-old mice overexpressing mutated α-synuclein (A53T mice) with tau oligomer-specific monoclonal antibody (TOMA) and a control antibody and assessed both behavioral and pathological phenotypes. RESULTS: We found that A53T mice treated with TOMA were protected from cognitive and motor deficits two weeks after a single injection. Levels of toxic tau oligomers were specifically decreased in the brains of TOMA-treated mice. Tau oligomer depletion also protected against dopamine and synaptic protein loss. CONCLUSION: These results indicate that targeting tau oligomers is beneficial for a mouse model of synucleinopathy and may be a viable therapeutic strategy for treating diseases in which tau and α-synuclein have a synergistic toxicity.


Assuntos
Anticorpos Monoclonais/farmacologia , Encéfalo/efeitos dos fármacos , Imunoterapia/métodos , alfa-Sinucleína , Proteínas tau/antagonistas & inibidores , Animais , Encéfalo/patologia , Humanos , Imunização Passiva , Camundongos , Camundongos Transgênicos , Doença de Parkinson/patologia
5.
Biol Psychiatry ; 84(7): 499-508, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29478699

RESUMO

BACKGROUND: The coexistence of α-synuclein and tau aggregates in several neurodegenerative disorders, including Parkinson's disease and Alzheimer's disease, raises the possibility that a seeding mechanism is involved in disease progression. METHODS: To further investigate the role of α-synuclein in the tau aggregation pathway, we performed a set of experiments using both recombinant and brain-derived tau and α-synuclein oligomers to seed monomeric tau aggregation in vitro and in vivo. Brain-derived tau oligomers were isolated from well-characterized cases of progressive supranuclear palsy (n = 4) and complexes of brain-derived α-synuclein/tau oligomers isolated from patients with Parkinson's disease (n = 4). The isolated structures were purified and characterized by standard biochemical methods, then injected into Htau mice (n = 24) to assess their toxicity and role in tau aggregation. RESULTS: We found that α-synuclein induced a distinct toxic tau oligomeric strain that avoids fibril formation. In vivo, Parkinson's disease brain-derived α-synuclein/tau oligomers administered into Htau mouse brains accelerated endogenous tau oligomer formation concurrent with increasing cell loss. CONCLUSIONS: Our findings provide evidence, for the first time, that α-synuclein enhances the harmful effects of tau, thus contributing to disease progression.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Doença de Parkinson/metabolismo , Paralisia Supranuclear Progressiva/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animais , Progressão da Doença , Humanos , Camundongos , Proteínas Recombinantes
6.
Aging Dis ; 8(3): 257-266, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28580182

RESUMO

The importance of vascular contributions to cognitive impairment and dementia (VCID) associated with Alzheimer's disease (AD) and related neurodegenerative diseases is increasingly recognized, however, the underlying mechanisms remain obscure. There is growing evidence that in addition to Aß deposition, accumulation of hyperphosphorylated oligomeric tau contributes significantly to AD etiology. Tau oligomers are toxic and it has been suggested that they propagate in a "prion-like" fashion, inducing endogenous tau misfolding in cells. Their role in VCID, however, is not yet understood. The present study was designed to determine the severity of vascular deposition of oligomeric tau in the brain in patients with AD and related tauopathies, including dementia with Lewy bodies (DLB) and progressive supranuclear palsy (PSP). Further, we examined a potential link between vascular deposition of fibrillar Aß and that of tau oligomers in the Tg2576 mouse model. We found that tau oligomers accumulate in cerebral microvasculature of human patients with AD and PSP, in association with vascular endothelial and smooth muscle cells. Cerebrovascular deposition of tau oligomers was also found in DLB patients. We also show that tau oligomers accumulate in cerebral microvasculature of Tg2576 mice, partially in association with cerebrovascular Aß deposits. Thus, our findings add to the growing evidence for multifaceted microvascular involvement in the pathogenesis of AD and other neurodegenerative diseases. Accumulation of tau oligomers may represent a potential novel mechanism by which functional and structural integrity of the cerebral microvessels is compromised.

7.
Ann Clin Transl Neurol ; 4(4): 226-235, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28382304

RESUMO

OBJECTIVE: With an increasing incidence of Alzheimer's disease (AD) and neurodegenerative tauopathies, there is an urgent need to develop reliable biomarkers for the diagnosis and monitoring of the disease, such as the recently discovered toxic tau oligomers. Here, we aimed to demonstrate the presence of tau oligomers in the cerebrospinal fluid (CSF) of patients with cognitive deficits, and to determine whether tau oligomers could serve as a potential biomarker for AD. METHODS: A multicentric collaborative study involving a double-blinded analysis with a total of 98 subjects with moderate to severe AD (N = 41), mild AD (N = 31), and nondemented control subjects (N = 26), and two pilot studies of 33 total patients with AD (N = 19) and control (N = 14) subjects were performed. We carried out biochemical assays to measure oligomeric tau from CSF of these patients with various degrees of cognitive impairment as well as cognitively normal controls. RESULTS: Using a highly reproducible indirect ELISA method, we found elevated levels of tau oligomers in AD patients compared to age-matched controls. Western blot analysis confirmed the presence of oligomeric forms of tau in CSF. In addition, the ratio of oligomeric to total tau increased in the order: moderate to severe AD, mild AD, and controls. CONCLUSION: These assays are suitable for the analysis of human CSF samples. These results here suggest that CSF tau oligomer measurements could be optimized and added to the panel of CSF biomarkers for the accurate and early detection of AD.

8.
Front Aging Neurosci ; 9: 83, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28420982

RESUMO

Aging has long been considered as the main risk factor for several neurodegenerative disorders including a large group of diseases known as tauopathies. Even though neurofibrillary tangles (NFTs) have been examined as the main histopathological hallmark, they do not seem to play a role as the toxic entities leading to disease. Recent studies suggest that an intermediate form of tau, prior to NFT formation, the tau oligomer, is the true toxic species. However, the mechanisms by which tau oligomers trigger neurodegeneration remain unknown. This review summarizes recent findings regarding the role of tau oligomers in disease, including release from cells, propagation from affected to unaffected brain regions, uptake into cells, and toxicity via mitochondrial dysfunction. A greater understanding of tauopathies may lead to future advancements in regards to prevention and treatment.

9.
Neuromolecular Med ; 19(1): 161-174, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27664052

RESUMO

Ataxia-telangiectasia is a multisystemic disease with severe neurological affectation, immunodeficiency and telangiectasia. The disorder is caused by alterations in the ATM gene, whose size and complexity make molecular diagnosis difficult. We designed a target-enrichment next-generation sequencing strategy to characterize 28 patients from several regions of Spain. This approach allowed us to identify gene variants affecting function in 54 out of the 56 alleles analyzed, although the two unresolved alleles belong to brothers. We found 28 ATM gene mutations, of which 10 have not been reported. A total of 171 gene variants not affecting function were also found, of which 22 are reported to predispose to disease. Interestingly, all Roma (Spanish Gypsies) patients are homozygous for the same mutation and share the H3 ATM haplotype, which is strong evidence of a founder effect in this population. In addition, we generated a panel of 27 primary T cell lines from A-T patients, which revealed significant expression of ATM in two patients and traces of the protein in nine more. None of them retained residual ATM activity, and almost all T cell lines show increased or intermediate radiosensitivity.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/etnologia , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sequência de Bases , Linhagem Celular , Códon sem Sentido , Ensaio de Unidades Formadoras de Colônias , Análise Mutacional de DNA , Efeito Fundador , Mutação da Fase de Leitura , Haplótipos/genética , Humanos , Fosforilação , Polimorfismo de Nucleotídeo Único , Processamento de Proteína Pós-Traducional , Roma (Grupo Étnico)/genética , Alinhamento de Sequência , Análise de Sequência de DNA/métodos , Deleção de Sequência , Espanha/epidemiologia , Linfócitos T/metabolismo , Linfócitos T/patologia
10.
J Alzheimers Dis ; 55(3): 1083-1099, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27716675

RESUMO

It is well-established that inflammation plays an important role in Alzheimer's disease (AD) and frontotemporal lobar dementia (FTLD). Inflammation and synapse loss occur in disease prior to the formation of larger aggregates, but the contribution of tau to inflammation has not yet been thoroughly investigated. Tau pathologically aggregates to form large fibrillar structures known as tangles. However, evidence suggests that smaller soluble aggregates, called oligomers, are the most toxic species and form prior to tangles. Furthermore, tau oligomers can spread to neighboring cells and between anatomically connected brain regions. In addition, recent evidence suggests that inspecting the retina may be a window to brain pathology. We hypothesized that there is a relationship between tau oligomers and inflammation, which are hallmarks of early disease. We conducted immunofluorescence and biochemical analyses on tauopathy mice, FTLD, and AD subjects. We showed that oligomers co-localize with astrocytes, microglia, and HMGB1, a pro-inflammatory cytokine. Additionally, we show that tau oligomers are present in the retina and are associated with inflammatory cells suggesting that the retina may be a valid non-invasive biomarker for brain pathology. These results suggest that there may be a toxic relationship between tau oligomers and inflammation. Therefore, the ability of tau oligomers to spread may initiate a feed-forward cycle in which tau oligomers induce inflammation, leading to neuronal damage, and thus more inflammation. Further mechanistic studies are warranted in order to understand this relationship, which may have critical implications for improving the treatment of tauopathies.


Assuntos
Encefalite/etiologia , Doenças Neurodegenerativas/complicações , Retinite/etiologia , Retinite/metabolismo , Proteínas tau/metabolismo , Fatores Etários , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/metabolismo , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteína HMGB1/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Doenças Neurodegenerativas/genética , Neurônios/metabolismo , Neurônios/patologia , Retinite/patologia , Proteínas tau/genética
11.
J Neurotrauma ; 33(22): 2034-2043, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-26729399

RESUMO

Tau aggregation is a pathological feature of numerous neurodegenerative disorders and has also been shown to occur under certain conditions of traumatic brain injury (TBI). Currently, no effective treatments exist for the long-term effects of TBI. In some cases, TBI not only induces cognitive changes immediately post-injury, but also leads to increased incidence of neurodegeneration later in life. Growing evidence from our lab and others suggests that the oligomeric forms of tau initiate the onset and spread of neurodegenerative tauopathies. Previously, we have shown increased levels of brain-derived tau oligomers in autopsy samples from patients diagnosed with Alzheimer's disease. We have also shown similar increases in tau oligomers in animal models of neurodegenerative diseases and TBI. In the current study, we evaluated the presence of tau oligomers in blast-induced TBI. To test the direct impact of TBI-derived tau oligomer toxicity, we isolated tau oligomers from brains of rats that underwent either a blast- or a fluid percussion injury-induced TBI. Oligomers were characterized biochemically and morphologically and were then injected into hippocampi of mice overexpressing human tau (Htau). Mice were cognitively evaluated and brains were collected for immunological analysis after testing. We found that tau oligomers form as a result of brain injury in two different models of TBI. Additionally, these oligomers accelerated onset of cognitive deficits when injected into brains of Htau mice. Tau oligomer levels increased in the hippocampal injection sites and cerebellum, suggesting that tau oligomers may be responsible for seeding the spread of pathology post-TBI. Our results suggest that tau oligomers play an important role in the toxicity underlying TBI and may be a viable therapeutic target.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Proteínas tau/biossíntese , Proteínas tau/toxicidade , Animais , Disfunção Cognitiva/induzido quimicamente , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Técnicas Estereotáxicas/instrumentação , Proteínas tau/administração & dosagem
12.
Front Cell Neurosci ; 9: 464, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26696824

RESUMO

Alzheimer's disease (AD) is a progressive disorder in which the most noticeable symptoms are cognitive impairment and memory loss. However, the precise mechanism by which those symptoms develop remains unknown. Of note, neuronal loss occurs at sites where synaptic dysfunction is observed earlier, suggesting that altered synaptic connections precede neuronal loss. The abnormal accumulation of amyloid-ß (Aß) and tau protein is the main histopathological feature of the disease. Several lines of evidence suggest that the small oligomeric forms of Aß and tau may act synergistically to promote synaptic dysfunction in AD. Remarkably, tau pathology correlates better with the progression of the disease than Aß. Recently, a growing number of studies have begun to suggest that missorting of tau protein from the axon to the dendrites is required to mediate the detrimental effects of Aß. In this review we discuss the novel findings regarding the potential mechanisms by which tau oligomers contribute to synaptic dysfunction in AD.

13.
ACS Chem Neurosci ; 6(12): 1981-9, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26448453

RESUMO

Amyloid-beta (Aß) oligomers have emerged as the most toxic species in Alzheimer's disease (AD) and other amyloid pathologies. Also, Aß-42 peptide is more aggregation-prone compared to other Aß isoforms. Thus, we synthesized a small peptide of repeated sequence containing the last three amino acids, Val-40, Ile-41, and Ala-42 of Aß-42 that was subsequently aggregated and used to generate a novel antibody, VIA. In this study, we examined human AD and Tg2576 mouse brain samples using VIA in combination with other amyloid-specific antibodies and confirmed the specificity of VIA to oligomeric Aß-42. Moreover, we found that VIA does not recognize classic amyloid plaques composed of fibrillar Aß or Aß-40 ex vivo. Since VIA recognizes a distinct epitope specific to Aß-42 oligomers, it may have broad use for examining the accumulation of these oligomers in AD and other neurodegenerative diseases. VIA may also be used in immunotherapy studies to prevent neurodegenerative effects associated with Aß-42 oligomers.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Anticorpos/farmacologia , Encéfalo/metabolismo , Fatores Etários , Precursor de Proteína beta-Amiloide/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Neuroblastoma/patologia , Neuroimagem , Presenilina-1/genética , Ligação Proteica/efeitos dos fármacos
14.
J Neurosci ; 35(12): 4857-68, 2015 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-25810517

RESUMO

In Alzheimer's disease (AD), the pathological accumulation of tau appears to be a downstream effect of amyloid ß protein (Aß). However, the relationship between these two proteins and memory loss is unclear. In this study, we evaluated the specific removal of pathological tau oligomers in aged Tg2576 mice by passive immunotherapy using tau oligomer-specific monoclonal antibody. Removal of tau oligomers reversed memory deficits and accelerated plaque deposition in the brain. Surprisingly, Aß*56 levels decreased, suggesting a link between tau and Aß oligomers in the promotion of cognitive decline. The results suggest that tau oligomerization is not only a consequence of Aß pathology but also a critical mediator of the toxic effects observed afterward in AD. Overall, these findings support the potential of tau oligomers as a therapeutic target for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais/uso terapêutico , Proteínas tau/metabolismo , Envelhecimento/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Masculino , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/imunologia , Placa Amiloide/metabolismo , Proteínas tau/imunologia
15.
Biol Psychiatry ; 78(10): 672-83, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25676491

RESUMO

BACKGROUND: Aberrant accumulation of α-synuclein constitutes inclusion bodies that are considered a characteristic feature of a group of neurological disorders described as synucleinopathies. Often, multiple disease-causing proteins overlap within a given disease pathology. An emerging body of research focuses on the oligomeric populations of various pathogenic proteins, considering them as the culprits causing neuronal damage and degeneration. To this end, the use of conformation-specific antibodies has proven to be an effective tool. Previous work from our laboratory and others has shown that oligomeric entities of α-synuclein and tau accumulate in their respective diseases, but their interrelationship at this higher order has yet to be shown in synucleinopathies. METHODS: Here, we used two novel conformation-specific antibodies, F8H7 and Syn33, which recognize α-synuclein oligomers and were developed in our laboratory. We investigated brain tissue from five of each Parkinson's disease and dementia with Lewy bodies patients by performing biophysical and biochemical assays using these antibodies, in addition to the previously characterized anti-tau oligomer antibody T22. RESULTS: We demonstrate that in addition to the deposition of oligomeric α-synuclein, tau oligomers accumulate in these diseased brains compared with control brains. Moreover, we observed that oligomers of tau and α-synuclein exist in the same aggregates, forming hybrid oligomers in these patients' brains. CONCLUSIONS: In addition to the deposition of tau oligomers, our results also provide compelling evidence of co-occurrence of α-synuclein and tau into their most toxic forms, i.e., oligomers suggesting that these species interact and influence each other's aggregation via an interface in synucleinopathies.


Assuntos
Lobo Frontal/metabolismo , Doença por Corpos de Lewy/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Anticorpos , Ensaio de Imunoadsorção Enzimática , Feminino , Lobo Frontal/patologia , Humanos , Imuno-Histoquímica , Doença por Corpos de Lewy/patologia , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Doença de Parkinson/patologia , alfa-Sinucleína/imunologia , Proteínas tau/imunologia
16.
Int J Biol Macromol ; 72: 117-24, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25109455

RESUMO

We have conducted a thorough study of the exopolysaccharide (EPS) produced by strain HK30 of Halomonas stenophila, which we have named haloglycan. This strain was chosen during an ongoing research programme aimed at finding novel exopolysaccharide-producing halophilic bacteria in unexplored hypersaline habitats. Strain HK30 was isolated from a saline-wetland in Brikcha (Morocco) and identified as belonging to the species H. stenophila. It produced EPS mainly during the exponential growth phase and to a lesser extent during the stationary phase. Culture parameters influenced both bacterial growth and EPS production, EPS yield always being directly related to the quantity of biomass. Under optimum culture conditions, strain HK30 produced 3.89 g of EPS per litre of medium. The polymer was a sulphated heteropolysaccharide composed of two fractions, with molecular masses of 8.2 × 10(4) and 1.4 × 10(6). The crude EPS contained 44 ± 0.1% w/w carbohydrates and the following monosaccharide composition: glucose (24 ± 1.73), glucuronic acid (7.5 ± 0.37), mannose (5.5 ± 0.17), fucose (4.5 ± 0.36), galactose (1.2 ± 0.17) and rhamnose (1 ± 0.05) (%, w/w). It produced solutions of high viscosity and pseudoplastic behaviour that showed interesting flocculating and emulsifying activities and was also involved in forming biofilm.


Assuntos
Halomonas/química , Polissacarídeos Bacterianos/biossíntese , Biofilmes/crescimento & desenvolvimento , Emulsões/química , Floculação , Genes Bacterianos , Cinética , Peso Molecular , Filogenia , Polissacarídeos Bacterianos/ultraestrutura , RNA Ribossômico 16S/genética , Reologia , Soluções , Viscosidade
17.
Neurobiol Dis ; 71: 14-23, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25134727

RESUMO

Alzheimer's disease is a complex disease characterized by overlapping phenotypes with different neurodegenerative disorders. Oligomers are considered the most toxic species in amyloid pathologies. We examined human AD brain samples using an anti-oligomer antibody generated in our laboratory and detected potential hybrid oligomers composed of amyloid-ß, prion protein, α-synuclein, and TDP-43 phosphorylated at serines 409 and 410. These data and in vitro results suggest that Aß oligomer seeds act as a template for the aggregation of other proteins and generate an overlapping phenotype with other neuronal disorders. Furthermore, these results could explain why anti-amyloid-ß therapy has been unsuccessful.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Angiopatia Amiloide Cerebral/etiologia , Proteínas de Ligação a DNA/metabolismo , Lobo Frontal/metabolismo , Fragmentos de Peptídeos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Feminino , Lobo Frontal/patologia , Humanos , Imageamento Tridimensional , Masculino , Camundongos , Neuroblastoma/patologia , Neuroimagem , Príons/metabolismo , alfa-Sinucleína/metabolismo
18.
ACS Chem Neurosci ; 5(9): 752-69, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25075869

RESUMO

Neurodegenerative disease is one of the greatest health concerns today and with no effective treatment in sight, it is crucial that researchers find a safe and successful therapeutic. While neurofibrillary tangles are considered the primary tauopathy hallmark, more evidence continues to come to light to suggest that soluble, intermediate tau aggregates--tau oligomers--are the most toxic species in disease. These intermediate tau species may also be responsible for the spread of pathology, suggesting that oligomeric tau may be the best therapeutic target. Here, we summarize results for the modulation of tau by molecular chaperones, small molecules and aggregation inhibitors, post-translational modifications, immunotherapy, other techniques, and future directions.


Assuntos
Doenças Neurodegenerativas/terapia , Proteínas tau/metabolismo , Animais , Anticorpos/uso terapêutico , Humanos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/imunologia
19.
Acta Neuropathol Commun ; 2: 56, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24887264

RESUMO

Pathological aggregation of the microtubule-associated protein tau and subsequent accumulation of neurofibrillary tangles (NFTs) or other tau-containing inclusions are defining histopathological features of many neurodegenerative diseases, which are collectively known as tauopathies. Due to conflicting results regarding a correlation between the presence of NFTs and disease progression, the mechanism linking pathological tau aggregation with cell death is poorly understood. An emerging view is that NFTs are not the toxic entity in tauopathies; rather, tau intermediates between monomers and NFTs are pathogenic. Several proteins associated with neurodegenerative diseases, such as ß-amyloid (Aß) and α-synuclein, have the tendency to form pore-like amyloid structures (annular protofibrils, APFs) that mimic the membrane-disrupting properties of pore-forming protein toxins. The present study examined the similarities of tau APFs with other tau amyloid species and showed for the first time the presence of tau APFs in brain tissue from patients with progressive supranuclear palsy (PSP) and dementia with Lewy bodies (DLB), as well as in the P301L mouse model, which overexpresses mutated tau. Furthermore, we found that APFs are preceded by tau oligomers and do not go on to form NFTs, evading fibrillar fate. Collectively, our results demonstrate that in vivo APF formation depends on mutations in tau, phosphorylation levels, and cell type. These findings establish the pathological significance of tau APFs in vivo and highlight their suitability as therapeutic targets for several neurodegenerative tauopathies.


Assuntos
Encéfalo/metabolismo , Doença por Corpos de Lewy/patologia , Paralisia Supranuclear Progressiva/patologia , Proteínas tau/química , Proteínas tau/metabolismo , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Estruturas Celulares/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteína Básica da Mielina/metabolismo , Neuroblastoma/patologia , Fosforilação/fisiologia
20.
J Alzheimers Dis ; 40 Suppl 1: S97-S111, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24603946

RESUMO

Neurodegenerative disease is one of the greatest health crises in the world and as life expectancy rises, the number of people affected will continue to increase. The most common neurodegenerative disease, Alzheimer's disease, is a tauopathy, characterized by the presence of aggregated tau, namely in the form of neurofibrillary tangles. Historically, neurofibrillary tangles have been considered the main tau species of interest in Alzheimer's disease; however, we and others have shown that tau oligomers may be the most toxic form and the species responsible for the spread of pathology. We developed a novel anti-tau oligomer-specific mouse monoclonal antibody (TOMA) and investigated the potential of anti-tau oligomer passive immunization in preventing the toxicity of tau pathology in Htau mice. We injected pure brain-derived tau oligomers intracerebrally in 3-month-old wild-type and Htau mice and investigated the protective effects of a single 60 µg TOMA injection when compared to the same dose of non-specific IgG and found that TOMA conferred protection against the accumulation of tau oligomers and cognitive deficits for up to 1 month after treatment. Additionally, we injected pure brain-derived tau oligomers intracerebrally in 3-month-old wild-type and Htau mice and treated animals with biweekly injections of 60 µg TOMA or non-specific IgG. We found that long-term administration of TOMA was effective as a preventative therapy, inhibiting oligomeric tau and preserving memory function. These results support the critical role of oligomeric tau in disease progression and validate tau oligomers as a potential drug target.


Assuntos
Transtornos Cognitivos/prevenção & controle , Imunização Passiva , Tauopatias/terapia , Proteínas tau/imunologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/imunologia , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Camundongos Transgênicos , Tauopatias/complicações , Tauopatias/imunologia , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...