Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nutr Res Rev ; 36(2): 471-483, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36156184

RESUMO

As we continue to elucidate the mechanisms underlying age-related brain diseases, the reductionist strategy in nutrition­brain function research has focused on establishing the impact of individual foods. However, the biological processes connecting diet and cognition are complex. Therefore, consideration of a combination of nutritional compounds may be most efficacious. One barrier to establishing the efficacy of multi-nutrient interventions is that the area lacks an established set of evidence-based guidelines for studying their effect on brain health. This review is an output of the International Life Sciences Institute (ILSI) Europe. A multi-disciplinary expert group was assembled with the aim of developing a set of considerations to guide research into the effects of multi-nutrient combinations on brain functions. Consensus recommendations converged on six key issues that should be considered to advance research in this area: (1) establish working mechanisms of the combination and contributions of each individual compound; (2) validate the relevance of the mechanisms for the targeted human condition; (3) include current nutrient status, intake or dietary pattern as inclusion/exclusion criteria in the study design; (4) select a participant population that is clinically and biologically appropriate for all nutritional components of the combination; (5) consider a range of cognitive outcomes; (6) consider the limits of reductionism and the 'gold standard' randomised controlled trial. These guiding principles will enhance our understanding of the interactive/complementary activities of dietary components, thereby strengthening the evidence base for recommendations aimed at delaying cognitive decline.


Assuntos
Envelhecimento Cognitivo , Nutrientes , Humanos , Alimentos , Encéfalo , Cognição , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
Arterioscler Thromb Vasc Biol ; 39(9): e195-e207, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31315437

RESUMO

OBJECTIVE: The ApoE (apolipoprotein) allele epsilon 4 is a major genetic risk factor for Alzheimer disease, cardiovascular disorders, and stroke, indicating that it significantly impacts cerebral and vascular systems. However, very little is known about how APOE genotype affects brain endothelial cells, which form a network of tight junctions to regulate communication between the brain and circulating blood factors. Approach and Results: Here, we present a novel model of endothelial dysfunction using isogenic human induced pluripotent stem cell-derived cells harboring different alleles of the APOE gene, specifically ApoE 3/3, 3/4, and 4/4. We show for the first time that ApoE4 expression by endothelial cells is sufficient to cause a toxic gain of cellular dysfunction. Using RNAseq, we found significant effects of ApoE4 on signaling pathways involved in blood coagulation and barrier function. These changes were associated with altered cell function, including increased binding of platelets to ECs with the 3/4 or 4/4 genotype. ApoE4-positive cells exhibited a proinflammatory state and prothrombotic state, evidenced by higher secretion of Aß (amyloid-ß) 40 and 42, increased release of cytokines, and overexpression of the platelet-binding protein VWF (vonWillebrand factor). Immunohistochemistry of human brain Alzheimer disease brains also showed increased VWF expression with the ApoE4/4 genotype. Finally, pharmacological inhibition of inflammation in ECs by celastrol rescued overexpression of VWF in cells expressing ApoE4. CONCLUSIONS: These cells provide novel insight into ApoE4-mediated endothelial dysfunction and provide a new platform to test potential therapies for vascular disorders.


Assuntos
Apolipoproteína E4/fisiologia , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Doença de Alzheimer/etiologia , Apolipoproteína E4/genética , Barreira Hematoencefálica , Genótipo , Humanos , Corpos de Weibel-Palade/fisiologia , Fator de von Willebrand/genética , Fator de von Willebrand/fisiologia
3.
EMBO J ; 34(24): 3028-41, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26538322

RESUMO

In Alzheimer's disease and tauopathies, tau protein aggregates into neurofibrillary tangles that progressively spread to synaptically connected brain regions. A prion-like mechanism has been suggested: misfolded tau propagating through the brain seeds neurotoxic aggregation of soluble tau in recipient neurons. We use transgenic mice and viral tau expression to test the hypotheses that trans-synaptic tau propagation, aggregation, and toxicity rely on the presence of endogenous soluble tau. Surprisingly, mice expressing human P301Ltau in the entorhinal cortex showed equivalent tau propagation and accumulation in recipient neurons even in the absence of endogenous tau. We then tested whether the lack of endogenous tau protects against misfolded tau aggregation and toxicity, a second prion model paradigm for tau, using P301Ltau-overexpressing mice with severe tangle pathology and neurodegeneration. Crossed onto tau-null background, these mice had similar tangle numbers but were protected against neurotoxicity. Therefore, misfolded tau can propagate across neural systems without requisite templated misfolding, but the absence of endogenous tau markedly blunts toxicity. These results show that tau does not strictly classify as a prion protein.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas tau/genética , Animais , Células Cultivadas , Córtex Entorrinal/citologia , Córtex Entorrinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Neurônios/metabolismo , Proteínas tau/deficiência , Proteínas tau/metabolismo
4.
Synapse ; 71(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28196395

RESUMO

Synaptic dysfunction and loss is the strongest pathological correlate of cognitive decline in Alzheimer's disease (AD) with increasing evidence implicating neuropathological tau protein in this process. Despite the knowledge that tau spreads through defined synaptic circuits, it is currently unknown whether synapse loss occurs before the accumulation of tau or as a consequence. To address this, we have used array tomography to examine an rTgTauEC mouse model expressing a P301L human tau transgene and a transgene labeling cytoplasm red (tdTomato) and presynaptic terminals green (Synaptophysin-EGFP). All transgenes are restricted primarily to the entorhinal cortex using the neuropsin promotor to drive tTA expression. It has previously been shown that rTgTauEC mice exhibit neuronal loss in the entorhinal cortex and synapse density loss in the middle molecular layer (MML) of the dentate gyrus at 24 months of age. Here, we observed the density of tau-expressing and total presynapses, and the spread of tau into the postsynapse in the MML of 3-6, 9, and 18 month old red-green-rTgTauEC mice. We observe no loss of synapse density in the MML up to 18 months even in axons expressing tau. Despite the maintenance of synapse density, we see spread of human tau from presynaptic terminals to postsynaptic compartments in the MML at very early ages, indicating that the spread of tau through neural circuits is not due to the degeneration of axon terminals and is an early feature of the disease process.


Assuntos
Doença de Alzheimer/metabolismo , Neurônios/metabolismo , Terminações Pré-Sinápticas/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Axônios/metabolismo , Axônios/patologia , Morte Celular , Córtex Entorrinal/metabolismo , Córtex Entorrinal/patologia , Feminino , Masculino , Camundongos , Neurônios/patologia , Terminações Pré-Sinápticas/patologia , Proteínas tau/genética
5.
Neurobiol Dis ; 85: 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26459111

RESUMO

Expression of the frontotemporal dementia-related tau mutation, P301L, at physiological levels in adult mouse brain (KI-P301L mice) results in overt hypophosphorylation of tau and age-dependent alterations in axonal mitochondrial transport in peripheral nerves. To determine the effects of P301L tau expression in the central nervous system, we examined the kinetics of mitochondrial axonal transport and tau phosphorylation in primary cortical neurons from P301L knock-in (KI-P301L) mice. We observed a significant 50% reduction in the number of mitochondria in the axons of cortical neurons cultured from KI-P301L mice compared to wild-type neurons. Expression of murine P301L tau did not change the speed, direction of travel or likelihood of movement of mitochondria. Notably, the angle that defines the orientation of the mitochondria in the axon, and the volume of individual moving mitochondria, were significantly increased in neurons expressing P301L tau. We found that murine tau phosphorylation in KI-P301L mouse neurons was diminished and the ability of P301L tau to bind to microtubules was also reduced compared to tau in wild-type neurons. The P301L mutation did not influence the ability of murine tau to associate with membranes in cortical neurons or in adult mouse brain. We conclude that P301L tau is associated with mitochondrial changes and causes an early reduction in murine tau phosphorylation in neurons coupled with impaired microtubule binding of tau. These results support the association of mutant tau with detrimental effects on mitochondria and will be of significance for the pathogenesis of tauopathies.


Assuntos
Axônios/metabolismo , Mitocôndrias/metabolismo , Proteínas tau/metabolismo , Animais , Axônios/patologia , Membrana Celular/metabolismo , Membrana Celular/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Citosol/metabolismo , Citosol/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microtúbulos/metabolismo , Mitocôndrias/patologia , Mutação , Fosforilação , Ratos , Proteínas tau/genética
6.
EMBO Rep ; 14(4): 389-94, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23412472

RESUMO

Propagation of tau pathology is linked with progressive neurodegeneration, but the mechanism underlying trans-synaptic spread of tau is unknown. We show that stimulation of neuronal activity, or AMPA receptor activation, induces tau release from healthy, mature cortical neurons. Notably, phosphorylation of extracellular tau appears reduced in comparison with intracellular tau. We also find that AMPA-induced release of tau is calcium-dependent. Blocking pre-synaptic vesicle release by tetanus toxin and inhibiting neuronal activity with tetrodotoxin both significantly impair AMPA-mediated tau release. Tau secretion is therefore a regulatable process, dysregulation of which could lead to the spread of tau pathology in disease.


Assuntos
Neurônios/metabolismo , Proteínas tau/metabolismo , Potenciais de Ação , Animais , Células Cultivadas , Córtex Cerebral/citologia , Potássio/fisiologia , Cultura Primária de Células , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/metabolismo
7.
Hum Mol Genet ; 21(24): 5254-67, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22965877

RESUMO

Abnormal phosphorylation of the microtubule-associated protein tau in neurodegenerative disorders, including Alzheimer's disease (AD) and frontotemporal lobar degeneration, is associated with disrupted axonal transport and synaptic dysfunction ultimately manifesting as histopathological lesions of protein aggregates. Glycogen synthase kinase 3ß (GSK3ß) may be critical for the pathological hyperphosphorylation of tau. Here, we examined the role of the proteasome-associated protein Nedd8 ultimate buster 1 (NUB1) in the neuropathogenic phosphorylation and aggregation of tau. We reveal that NUB1 interacted with both tau and GSK3ß to disrupt their interaction, and abolished recruitment of GSK3ß to tau inclusions. Moreover, NUB1 reduced GSK3ß-mediated phosphorylation of tau and aggregation of tau in intracellular inclusions. Strikingly, NUB1 induced GSK3ß degradation. Deletion of the NUB1 ubiquitin-like (UBL) domain did not impair the interaction with tau and GSK3ß, and the ability to suppress the phosphorylation and aggregation of tau was not affected. However, the UBL motif was necessary for GSK3ß degradation. Deletion of the NUB1 ubiquitin-associated (UBA) domain abrogated the ability of NUB1 to interact with and degrade GSK3ß. Moreover, the UBA domain was required to suppress the aggregation of tau. Silencing of NUB1 in cells stabilized endogenous GSK3ß and exacerbated tau phosphorylation. Thus, we propose that NUB1, by regulating GSK3ß levels, modulates tau phosphorylation and aggregation, and is a key player in neurodegeneration associated with tau pathology. Moreover, NUB1 regulation of GSK3ß could modulate numerous signalling pathways in which GSK3ß is a centrally important effector.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas tau/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Imuno-Histoquímica , Imunoprecipitação , Fosforilação/genética , Fosforilação/fisiologia , Ligação Proteica/genética , Ligação Proteica/fisiologia , Interferência de RNA , Ratos , Proteínas tau/genética
8.
Acta Neuropathol ; 127(2): 257-70, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24271788

RESUMO

Neurofibrillary tangles (NFTs), a hallmark of Alzheimer's disease, are intracellular silver and thioflavin S-staining aggregates that emerge from earlier accumulation of phospho-tau in the soma. Whether soluble misfolded but nonfibrillar tau disrupts neuronal function is unclear. Here we investigate if soluble pathological tau, specifically directed to the entorhinal cortex (EC), can cause behavioral or synaptic deficits. We studied rTgTauEC transgenic mice, in which P301L mutant human tau overexpressed primarily in the EC leads to the development of tau pathology, but only rare NFT at 16 months of age. We show that the early tau lesions are associated with nearly normal performance in contextual fear conditioning, a hippocampal-related behavior task, but more robust changes in neuronal system activation as marked by Arc induction and clear electrophysiological defects in perforant pathway synaptic plasticity. Electrophysiological changes were likely due to a presynaptic deficit and changes in probability of neurotransmitter release. The data presented here support the hypothesis that misfolded and hyperphosphorylated tau can impair neuronal function within the entorhinal-hippocampal network, even prior to frank NFT formation and overt neurodegeneration.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Córtex Entorrinal/metabolismo , Terminações Pré-Sinápticas/fisiologia , Proteínas tau/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos/fisiologia , Córtex Entorrinal/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia
9.
Sci Adv ; 7(12)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33741591

RESUMO

Neuronal tau reduction confers resilience against ß-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases.


Assuntos
Doença de Alzheimer , Fatores de Transcrição , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Camundongos , Placa Amiloide/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética , Proteínas tau/genética , Proteínas tau/metabolismo
10.
Biochem Soc Trans ; 38(4): 1012-5, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658995

RESUMO

Tau is an abundant microtubule-associated protein which regulates the stability of the cytoskeleton. Tau binds microtubules directly through microtubule-binding domains in its C-terminus. However, tau is not only located in the cytosol of cells, but also associated with other intracellular domains, including the plasma membrane, suggesting that tau may have additional functions other than stabilizing the neuronal cytoskeleton. Localization of tau at the cell surface appears to be dependent on interactions of the N-terminal projection domain of tau. Furthermore, membrane-associated tau is dephosphorylated at serine/threonine residues, suggesting that the phosphorylation state of tau regulates its intracellular trafficking. Dephosphorylation of tau may increase the association of tau with trafficking proteins which target tau to the plasma membrane. Thus it is possible that the hyperphosphoryation of tau may contribute to the pathogenesis of Alzheimer's disease by promoting the formation of neurofibrillary tangles from cytosolic tau, and also by inhibiting additional tau functions through disruption of its targeting to the plasma membrane.


Assuntos
Membrana Celular/metabolismo , Proteínas tau/metabolismo , Proteínas tau/fisiologia , Animais , Membrana Celular/fisiologia , Humanos , Degeneração Neural , Fosforilação , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas tau/química
12.
Mol Neurobiol ; 54(5): 3428-3438, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27177549

RESUMO

Age remains the main risk factor for developing Alzheimer's disease (AD) although certain metabolic alterations, including prediabetes and type 2 diabetes (T2D), may also increase this risk. In order to understand this relationship, we have studied an AD-prediabetes mouse model (APP/PS1) with severe hyperinsulinemia induced by long-term high fat diet (HFD), and an AD-T2D model, generated by crossing APP/PS1 and db/db mice (APP/PS1xdb/db). In both, prediabetic and diabetic AD mice, we have analyzed underlying neuronal pathology and synaptic loss. At 26 weeks of age, when both pathologies were clearly established, we observed severe brain atrophy in APP/PS1xdb/db animals as well as cortical thinning, accompanied by increased caspase activity. Reduced senile plaque burden and elevated soluble Aß40 and 42 levels were observed in AD-T2D mice. Further assessment revealed a significant increase of neurite curvature in prediabetic-AD mice, and this effect was worsened in AD-T2D animals. Synaptic density loss, analyzed by array tomography, revealed a synergistic effect between T2D and AD, whereas an intermediate state was observed, once more, in prediabetic-AD mice. Altogether, our data suggest that early prediabetic hyperinsulinemia may exacerbate AD pathology, and that fully established T2D clearly worsens these effects. Therefore, it is feasible that early detection of prediabetic state and strict metabolic control could slow or delay progression of AD-associated neuropathological features.


Assuntos
Doença de Alzheimer/patologia , Diabetes Mellitus Tipo 2/patologia , Neurônios/patologia , Estado Pré-Diabético/patologia , Sinapses/patologia , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Atrofia/patologia , Caspases/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Camundongos Transgênicos , Neuritos/metabolismo , Neurônios/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Estado Pré-Diabético/complicações , Estado Pré-Diabético/metabolismo , Presenilina-1/metabolismo , Sinapses/metabolismo
13.
Cell Death Dis ; 8(3): e2671, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300838

RESUMO

The spatiotemporal transmission of pathological tau in the brain is characteristic of Alzheimer's disease. Release of both soluble and abnormal tau species from healthy neurons is increased upon stimulation of neuronal activity. It is not yet understood whether the mechanisms controlling soluble tau release from healthy neurons is the same as those involved in the spread of pathological tau species. To begin to understand these events, we have studied tau distribution and release using organotypic brain slice cultures. The slices were cultured from postnatal wild-type and 3xTg-AD mice for up to 1 month. Tau distribution in subcellular compartments was examined by western blotting, and tau release into culture medium was determined using a sensitive sandwich ELISA. We show here that 3xTg-AD cultures have an accelerated development of pathological tau abnormalities including the redistribution of tau to synaptic and membrane compartments. The 3xTg-AD slice cultures show elevated basal tau release relative to total tau when compared with wild-type cultures. However, tau release from 3xTg-AD slices cannot be further stimulated when neuronal activity is increased with potassium chloride. Moreover, we report that there is an increased pool of dephosphorylated membrane-associated tau in conditions where tau release is increased. These data suggest that there may be differential patterns of tau release when using integrated slice culture models of wild-type and transgenic mouse brain, although it will be important to determine the effect of tau overexpression for these findings. These results further increase our knowledge of the molecular mechanisms underlying tau release and propagation in neurodegenerative tauopathies.


Assuntos
Encéfalo/metabolismo , Membrana Celular/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Pareamento Cromossômico/fisiologia , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos/metabolismo , Neurônios/metabolismo , Fosforilação/fisiologia
14.
Neurobiol Aging ; 39: 1-18, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26923397

RESUMO

Hyperphosphorylation and fibrillar aggregation of the microtubule-associated protein tau are key features of Alzheimer's disease and other tauopathies. To investigate the involvement of tau phosphorylation in the pathological process, we generated a pair of complementary phosphomutant tau knockin mouse lines. One exclusively expresses phosphomimetic tau with 18 glutamate substitutions at serine and/or threonine residues in the proline-rich and first microtubule-binding domains to model hyperphosphorylation, whereas its phosphodefective counterpart has matched alanine substitutions. Consistent with expected effects of genuine phosphorylation, association of the phosphomimetic tau with microtubules and neuronal membranes is severely disrupted in vivo, whereas the phosphodefective mutations have more limited or no effect. Surprisingly, however, age-related mislocalization of tau is evident in both lines, although redistribution appears more widespread and more pronounced in the phosphomimetic tau knockin. Despite these changes, we found no biochemical or immunohistological evidence of pathological tau aggregation in mice of either line up to at least 2 years of age. These findings raise important questions about the role of tau phosphorylation in driving pathology in human tauopathies.


Assuntos
Tauopatias/genética , Tauopatias/patologia , Proteínas tau/metabolismo , Animais , Humanos , Camundongos Mutantes , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Agregação Patológica de Proteínas
15.
Acta Neuropathol Commun ; 4(1): 49, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27193083

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterised by neuropathological deposits of amyloid plaques and neurofibrillary tangles comprised of ß-amyloid and tau protein, respectively. In AD, tau becomes abnormally phosphorylated and aggregates to form intracellular deposits. However, the mechanisms by which tau exerts neurotoxicity in disease remain unclear. Recent studies have suggested that the presence of tau at synapses may indicate a role in neuronal signalling, which could be disrupted in pathological conditions. The non-receptor-associated tyrosine kinase fyn is located at the dendrite in neurons, where it was recently shown to interact with tau to stabilise receptor complexes at the post-synaptic density. Fyn also co-localises with tau in a proportion of neurons containing tau tangles in AD and fyn is also a tau kinase. Hence, tau-fyn interactions could play a pathogenic role in AD. Here we report the identification of critical proline residues, Pro213, Pro216, and Pro219, located within the fifth and sixth Pro-X-X-Pro motifs in the proline-rich region of tau, that are important for its binding to fyn. These residues in tau are flanked by numerous phosphorylation sites and therefore we investigated the relationship between fyn and the degree of tau phosphorylation in human post-mortem brain tissue. We found no difference in the amount of fyn present in control and AD brain. Notably, however, there was a significant correlation between fyn and phosphorylated tau at specific phospho-epitopes in control, but not in AD brain. Our results suggest that the pathological mechanisms underlying AD, that result in increased tau phosphorylation, may disrupt the physiological relationship between tau phosphorylation and fyn.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Motivos de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células CHO , Cricetulus , Escherichia coli , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação , Ligação Proteica , Domínios Proteicos
16.
J Mol Neurosci ; 27(1): 137-45, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16055952

RESUMO

Membrane phospholipids like phosphatidylcholine (PC) are required for cellular growth and repair, and specifically for synaptic function. PC synthesis is controlled by cellular levels of its precursor, cytidine-5'-diphosphate choline (CDP-choline), which is produced from cytidine triphosphate (CTP) and phosphocholine. In rat PC12 cells exogenous uridine was shown to elevate intracellular CDP-choline levels, by promoting the synthesis of uridine triphosphate (UTP), which was partly converted to CTP. In such cells uridine also enhanced the neurite outgrowth produced by nerve growth factor (NGF). The present study assessed the effect of dietary supplementation with uridine-5'-monophosphate disodium (UMP-2Na+, an additive in infant milk formulas) on striatal dopamine (DA) release in aged rats. Male Fischer 344 rats consumed either a control diet or one fortified with 2.5% UMP for 6 wk, ad libitum. In vivo microdialysis was then used to measure spontaneous and potassium (K+)-evoked DA release in the right striatum. Potassium (K+)-evoked DA release was significantly greater among UMP-treated rats, i.e., 341+/-21% of basal levels vs. 283+/-9% of basal levels in control rats (p<0.05); basal DA release was unchanged. In general, each animal's K+-evoked DA release correlated with its striatal DA content, measured postmortem. The levels of neurofilament-70 and neurofilament-M proteins, biomarkers of neurite outgrowth, increased to 182+/-25% (p<0.05) and 221+/-34% (p<0.01) of control values, respectively, with UMP consumption. Hence, UMP treatment not only enhances membrane phosphatide production but also can modulate two membrane-dependent processes, neurotransmitter release and neurite outgrowth, in vivo.


Assuntos
Envelhecimento/fisiologia , Dieta , Suplementos Nutricionais , Dopamina/metabolismo , Neuritos/metabolismo , Potássio/metabolismo , Uridina Monofosfato/administração & dosagem , Animais , Corpo Estriado/química , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Masculino , Microdiálise , Proteínas de Neurofilamentos/metabolismo , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Uridina Monofosfato/metabolismo
17.
Acta Neuropathol Commun ; 3: 14, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25853174

RESUMO

INTRODUCTION: In early stages of Alzheimer's disease (AD), neurofibrillary tangles (NFT) are largely restricted to the entorhinal cortex and medial temporal lobe. At later stages, when clinical symptoms generally occur, NFT involve widespread limbic and association cortices. At this point in the disease, amyloid plaques are also abundantly distributed in the cortex. This observation from human neuropathological studies led us to pose two alternative hypotheses: that amyloid in the cortex is permissive for the spread of tangles from the medial temporal lobe, or that these are co-occurring but not causally related events simply reflecting progression of AD pathology. RESULTS: We now directly test the hypothesis that cortical amyloid acts as an accelerant for spreading of tangles beyond the medial temporal lobe. We crossed rTgTauEC transgenic mice that demonstrate spread of tau from entorhinal cortex to other brain structures at advanced age with APP/PS1 mice, and examined mice with either NFTs, amyloid pathology, or both. We show that concurrent amyloid deposition in the cortex 1) leads to a dramatic increase in the speed of tau propagation and an extraordinary increase in the spread of tau to distal brain regions, and 2) significantly increases tau-induced neuronal loss. CONCLUSIONS: These data strongly support the hypothesis that cortical amyloid accelerates the spread of tangles throughout the cortex and amplifies tangle-associated neural system failure in AD.


Assuntos
Doença de Alzheimer/patologia , Amiloide/metabolismo , Encéfalo/patologia , Emaranhados Neurofibrilares/patologia , Neurônios/patologia , Proteínas tau/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Amiloide/toxicidade , Animais , Encéfalo/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Progressão da Doença , Hipocampo/patologia , Humanos , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Lobo Temporal/patologia
18.
Brain Res ; 951(1): 135-40, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12231467

RESUMO

Stimulation of serotonin receptor subtypes 5-HT(2A) or 5-HT(2C) in stably transfected 3T3 cells by dexnorfenfluramine (DEXNOR) or serotonin increases secretion of the APP metabolite APP(s). It is not known whether activation of these receptors can also affect APP metabolism in vivo. We examined the effects of a single intraperitoneal (i.p.) injection of DEXNOR on APP(s) levels in cerebrospinal fluid (CSF) of guinea pigs. These levels were significantly (P<0.05) increased by a single dose of DEXNOR (1-4 mg/kg); those of the APP metabolites Abeta(1-40) and Abeta(1-42) were unaffected. The DEXNOR-induced (1 mg/kg) increases in CSF APP(s) were suppressed by ritanserin (1 mg/kg) but not by ketanserin (2 mg/kg). When given alone, ritanserin did not affect CSF levels of APP(s), Abeta(1-40), or Abeta(1-42). Chronic treatment with DEXNOR for 9 days (1 mg/kg bid, i.p.) increased CSF APP(s) levels, measured 2 h after the last injection (P<0.05), and decreased those of CSF Abeta(1-42) (P<0.05). Neither hippocampal nor cortical levels of the APP holoprotein (APP(h)), nor body weight, were affected by DEXNOR. Chronic administration of mCPP (1-(m-chlorophenyl)piperazine) (2 mg/kg bid, i.p.), a 5-HT(2B/2C) agonist, for 9 days also increased CSF APP(s) levels (P<0.5) when measured 2 h after the drug's last administration; hippocampal and cortical APP(h) levels were unaffected. However, mCPP also caused a significant decrease in body weight gain. These data indicate that the pharmacological activation of 5-HT(2C) receptors can stimulate CSF APP(s) secretion and reduce Abeta production in vivo. Hence 5-HT(2C) receptors, which apparently are localized to the brain, may represent useful targets for the development of treatments for Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Norfenfluramina/farmacologia , Fragmentos de Peptídeos/biossíntese , Receptores de Serotonina/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/líquido cefalorraquidiano , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Encéfalo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Cobaias , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ketanserina/farmacologia , Masculino , Receptor 5-HT2C de Serotonina , Receptores de Serotonina/metabolismo , Ritanserina/farmacologia , Serotonina/metabolismo
19.
Neurosci Lett ; 362(2): 127-30, 2004 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15193769

RESUMO

We investigated the effects of prostaglandin E2 (PGE2) on amyloid precursor protein (APP) expression in cultured rat microglia. PGE2 treatment significantly increased the expression of APP holoprotein and was associated with an elevation in cyclic AMP (cAMP). Direct activation of adenylate cyclase with forskolin also increased APP expression. Co-treatment of microglia with PGE2 and the PKA inhibitor H-89 suppressed the overexpression of APP caused by PGE2 alone. The prostaglandin EP2 receptor is known to be positively coupled to cAMP production. Stimulation of the EP2 receptor with butaprost increased APP holoprotein, whereas co-incubation of the cells with PGE(2) and the EP2 receptor antagonist AH-6809 blocked the effect of PGE2 on APP expression. These data suggest that PGE2 is able to regulate the expression of APP, and that this effect may be mediated by the EP2 receptor and the cAMP signaling cascade.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Dinoprostona/farmacologia , Microglia/efeitos dos fármacos , Receptores de Prostaglandina E/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Microglia/metabolismo , Ratos , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2 , Xantonas/farmacologia
20.
Neuropharmacology ; 76 Pt A: 1-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24076336

RESUMO

Alzheimer's disease (AD) is characterized by brain deposition of amyloid plaques and tau neurofibrillary tangles along with steady cognitive decline. Although the mechanism by which AD pathogenesis occurs is unclear, accumulating evidence suggests that dysfunction and loss of synaptic connections may be an early event underlying disease progression. Profound synapse degeneration is observed in AD, and the density of these connections strongly correlates with cognitive ability. Initial investigations into AD-related synaptic changes focused on the toxic effects of amyloid. However, recent research suggests an emerging role for tau at the synapse. Even in the absence of tangles, mice overexpressing human tau display significant synaptic degeneration, suggesting that soluble, oligomeric tau is the synaptotoxic species. However, the localization of tau within synapses in both healthy and AD brains indicates that tau might play a role in normal synaptic function, which may be disrupted in disease. Tau is able to impact synaptic activity in several ways: studies show tau interacting directly with post-synaptic signaling complexes, regulating glutamatergic receptor content in dendritic spines, and influencing targeting and function of synaptic mitochondria. Early trials of tau-targeted immunotherapy reduce tau pathology and synapse loss, indicating that the toxic effects of tau may be reversible within a certain time frame. Understanding the role of tau in both normal and degenerating synapses is crucial for the development of therapeutic strategies designed to ameliorate synapse loss and prevent AD pathogenesis. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.


Assuntos
Doença de Alzheimer/fisiopatologia , Degeneração Neural/fisiopatologia , Proteínas tau/fisiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Mitocôndrias/metabolismo , Modelos Neurológicos , Degeneração Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Transmissão Sináptica/fisiologia , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA