Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(12): eadk9884, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38507480

RESUMEN

Molecular chaperones are protective in neurodegenerative diseases by preventing protein misfolding and aggregation, such as extracellular amyloid plaques and intracellular tau neurofibrillary tangles in Alzheimer's disease (AD). In addition, AD is characterized by an increase in astrocyte reactivity. The chaperone HSPB1 has been proposed as a marker for reactive astrocytes; however, its astrocytic functions in neurodegeneration remain to be elucidated. Here, we identify that HSPB1 is secreted from astrocytes to exert non-cell-autonomous protective functions. We show that in human AD brain, HSPB1 levels increase in astrocytes that cluster around amyloid plaques, as well as in the adjacent extracellular space. Moreover, in conditions that mimic an inflammatory reactive response, astrocytes increase HSPB1 secretion. Concomitantly, astrocytes and neurons can uptake astrocyte-secreted HSPB1, which is accompanied by an attenuation of the inflammatory response in reactive astrocytes and reduced pathological tau inclusions. Our findings highlight a protective mechanism in disease conditions that encompasses the secretion of a chaperone typically regarded as intracellular.


Asunto(s)
Enfermedad de Alzheimer , Astrocitos , Humanos , Astrocitos/metabolismo , Proteínas tau/metabolismo , Placa Amiloide/patología , Neuroprotección , Chaperonas Moleculares/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas de Choque Térmico/metabolismo
4.
Brain Behav Immun ; 114: 414-429, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37716378

RESUMEN

The purinoceptor P2X7R is a promising therapeutic target for tauopathies, including Alzheimer's disease (AD). Pharmacological inhibition or genetic knockdown of P2X7R ameliorates cognitive deficits and reduces pathological tau burden in mice that model aspects of tauopathy, including mice expressing mutant human frontotemporal dementia (FTD)-causing forms of tau. However, disagreements remain over which glial cell types express P2X7R and therefore the mechanism of action is unresolved. Here, we show that P2X7R protein levels increase in human AD post-mortem brain, in agreement with an upregulation of P2RX7 mRNA observed in transcriptome profiles from the AMP-AD consortium. P2X7R protein increases mirror advancing Braak stage and coincide with synapse loss. Using RNAScope we detect P2RX7 mRNA in microglia and astrocytes in human AD brain, including in the vicinity of senile plaques. In cultured microglia, P2X7R activation modulates the NLRP3 inflammasome pathway by promoting the formation of active complexes and release of IL-1ß. In astrocytes, P2X7R activates NFκB signalling and increases production of the cytokines CCL2, CXCL1 and IL-6 together with the acute phase protein Lcn2. To further explore the role of P2X7R in a disease-relevant context, we expressed wild-type or FTD-causing mutant forms of tau in mouse organotypic brain slice cultures. Inhibition of P2X7R reduces insoluble tau levels without altering soluble tau phosphorylation or synaptic localisation, suggesting a non-cell autonomous role of glial P2X7R on pathological tau aggregation. These findings support further investigations into the cell-type specific effects of P2X7R-targeting therapies in tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Demencia Frontotemporal , Tauopatías , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Microglía/metabolismo , ARN Mensajero/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/metabolismo
5.
Alzheimers Dement ; 19(12): 5970-5987, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37768001

RESUMEN

INTRODUCTION: Experimental models are essential tools in neurodegenerative disease research. However, the translation of insights and drugs discovered in model systems has proven immensely challenging, marred by high failure rates in human clinical trials. METHODS: Here we review the application of artificial intelligence (AI) and machine learning (ML) in experimental medicine for dementia research. RESULTS: Considering the specific challenges of reproducibility and translation between other species or model systems and human biology in preclinical dementia research, we highlight best practices and resources that can be leveraged to quantify and evaluate translatability. We then evaluate how AI and ML approaches could be applied to enhance both cross-model reproducibility and translation to human biology, while sustaining biological interpretability. DISCUSSION: AI and ML approaches in experimental medicine remain in their infancy. However, they have great potential to strengthen preclinical research and translation if based upon adequate, robust, and reproducible experimental data. HIGHLIGHTS: There are increasing applications of AI in experimental medicine. We identified issues in reproducibility, cross-species translation, and data curation in the field. Our review highlights data resources and AI approaches as solutions. Multi-omics analysis with AI offers exciting future possibilities in drug discovery.


Asunto(s)
Demencia , Enfermedades Neurodegenerativas , Humanos , Inteligencia Artificial , Reproducibilidad de los Resultados , Aprendizaje Automático
6.
Science ; 381(6656): 377-378, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37499007

RESUMEN

Tau quality control by tripartite motif 11 (TRIM11) protects neurons in mice.


Asunto(s)
Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteínas tau , Animales , Ratones
7.
Front Mol Biosci ; 8: 779240, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34778381

RESUMEN

Tau35 is a truncated form of tau found in human brain in a subset of tauopathies. Tau35 expression in mice recapitulates key features of human disease, including progressive increase in tau phosphorylation, along with cognitive and motor dysfunction. The appearance of aggregated tau suggests that Tau35 may have structural properties distinct from those of other tau species that could account for its pathological role in disease. To address this hypothesis, we performed a structural characterization of monomeric and aggregated Tau35 and compared the results to those of two longer isoforms, 2N3R and 2N4R tau. We used small angle X-ray scattering to show that Tau35, 2N3R and 2N4R tau all behave as disordered monomeric species but Tau35 exhibits higher rigidity. In the presence of the poly-anion heparin, Tau35 increases thioflavin T fluorescence significantly faster and to a greater extent than full-length tau, demonstrating a higher propensity to aggregate. By using atomic force microscopy, circular dichroism, transmission electron microscopy and X-ray fiber diffraction, we provide evidence that Tau35 aggregation is mechanistically and morphologically similar to previously reported tau fibrils but they are more densely packed. These data increase our understanding of the aggregation inducing properties of clinically relevant tau fragments and their potentially damaging role in the pathogenesis of human tauopathies.

9.
Brain Commun ; 2(1)2020.
Artículo en Inglés | MEDLINE | ID: mdl-32500121

RESUMEN

Polymorphisms associated with BIN1 confer the second greatest risk for developing late onset Alzheimer's disease. The biological consequences of this genetic variation are not fully understood, however BIN1 is a binding partner for tau. Tau is normally a highly soluble cytoplasmic protein, but in Alzheimer's disease tau is abnormally phosphorylated and accumulates at synapses to exert synaptotoxicity. The purpose of this study was to determine if alterations to BIN1 and tau in Alzheimer's disease promote the damaging redistribution of tau to synapses, as a mechanism by which BIN1 polymorphisms may increase risk of developing Alzheimer's disease. We show that BIN1 is lost from the cytoplasmic fraction of Alzheimer's disease cortex, and this is accompanied by the progressive mislocalization of phosphorylated tau to synapses. We confirmed proline 216 in tau as critical for tau interaction with the BIN1-SH3 domain and show that phosphorylation of tau disrupts this binding, suggesting that tau phosphorylation in Alzheimer's disease disrupts tau-BIN1 associations. Moreover, we show that BIN1 knockdown in rat primary neurons to mimic BIN1 loss in Alzheimer's disease brain, causes the damaging accumulation of phosphorylated tau at synapses and alterations in dendritic spine morphology. We also observed reduced release of tau from neurons upon BIN1 silencing, suggesting that BIN1 loss disrupts the function of extracellular tau. Together, these data indicate that polymorphisms associated with BIN1 that reduce BIN1 protein levels in the brain likely act synergistically with increased tau phosphorylation to increase risk of Alzheimer's disease by disrupting cytoplasmic tau-BIN1 interactions, promoting the damaging mis-sorting of phosphorylated tau to synapses to alter synapse structure, and by reducing the release of physiological forms of tau to disrupt tau function.

11.
Acta Neuropathol Commun ; 7(1): 2, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30606258

RESUMEN

Human tauopathies including Alzheimer's disease, progressive supranuclear palsy and related disorders, are characterized by deposition of pathological forms of tau, synaptic dysfunction and neuronal loss. We have previously identified a pathogenic C-terminal tau fragment (Tau35) that is associated with human tauopathy. However, it is not known how tau fragmentation affects critical molecular processes in cells and contributes to impaired physiological function. Chinese hamster ovary (CHO) cells and new CHO cell lines stably expressing Tau35 or full-length human tau were used to compare the effects of disease-associated tau cleavage on tau function and signaling pathways. Western blots, microtubule-binding assays and immunofluorescence labeling were used to examine the effects of Tau35 on tau function and on signaling pathways in CHO cells. We show that Tau35 undergoes aberrant phosphorylation when expressed in cells. Although Tau35 contain the entire microtubule-binding region, the lack of the amino terminal half of tau results in a marked reduction in microtubule binding and defective microtubule organization in cells. Notably, Tau35 attenuates insulin-mediated activation of Akt and a selective inhibitory phosphorylation of glycogen synthase kinase-3. Moreover, Tau35 activates ribosomal protein S6 kinase beta-1 signaling and the unfolded protein response, leading to insulin resistance in cells. Tau35 has deleterious effects on signaling pathways that mediate pathological changes and insulin resistance, suggesting a mechanism through which N-terminal cleavage of tau leads to the development and progression of tau pathology in human tauopathy. Our findings highlight the importance of the N-terminal region of tau for its normal physiological function. Furthermore, we show that pathogenic tau cleavage induces tau phosphorylation, resulting in impaired microtubule binding, disruption of insulin signaling and activation of the unfolded protein response. Since insulin resistance is a feature of several tauopathies, this work suggests new potential targets for therapeutic intervention.


Asunto(s)
Insulina/metabolismo , Microtúbulos/metabolismo , Respuesta de Proteína Desplegada/fisiología , Proteínas tau/metabolismo , Animales , Células CHO , Cricetulus , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
12.
Adv Exp Med Biol ; 1184: 105-112, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32096032

RESUMEN

The microtubule-associated protein tau has been identified in several intraneuronal compartments, including in association with synapses. In Alzheimer's disease, frontotemporal dementia and related tauopathies, highly phosphorylated tau accumulates as intraneuronal protein aggregates that are likely responsible for the demise of neurons and the subsequent progressive cognitive decline. However, the molecular mechanisms underlying such tau-mediated damage in the tauopathies is not fully understood. Tauopathy induces loss of synapses, which is one of the earliest structural correlates of cognitive dysfunction and disease progression. Notably, altered post-translational modifications of tau, including increased phosphorylation and acetylation, augment the mislocalisation of tau to synapses, impair synaptic vesicle release and might influence the activity-dependent release of tau from neurons. Thus, disease-associated accumulation of modified tau at the synapse adversely affects critical neuronal processes that are linked to neuronal activity and synaptic function. These findings emphasise the importance of gaining a comprehensive understanding of the diverse roles of tau at distinct intraneuronal locations. An improved knowledge of the impact of synaptic tau under physiological and pathological conditions and how tau localisation impacts on neuronal function will provide valuable insights that may lead to the development of new therapies for the tauopathies.


Asunto(s)
Sinapsis/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Neuronas/metabolismo , Neuronas/patología , Sinapsis/patología , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/química
13.
Acta Neuropathol Commun ; 6(1): 65, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30037345

RESUMEN

Tau is normally a highly soluble phosphoprotein found predominantly in neurons. Six different isoforms of tau are expressed in the adult human CNS. Under pathological conditions, phosphorylated tau aggregates are a defining feature of neurodegenerative disorders called tauopathies. Recent findings have suggested a potential role of the gut-brain axis in CNS homeostasis, and therefore we set out to examine the isoform profile and phosphorylation state of tau in the enteric nervous system (ENS) under physiological conditions and in tauopathies. Surgical specimens of human colon from controls, Parkinson's disease (PD) and progressive supranuclear palsy (PSP) patients were analyzed by Western Blot and immunohistochemistry using a panel of anti-tau antibodies. We found that adult human ENS primarily expresses two tau isoforms, localized in the cell bodies and neuronal processes. We did not observe any difference in the enteric tau isoform profile and phosphorylation state between PSP, PD and control subjects. The htau mouse model of tauopathy also expressed two main isoforms of human tau in the ENS, and there were no apparent differences in ENS tau localization or phosphorylation between wild-type and htau mice. Tau in both human and mouse ENS was found to be phosphorylated but poorly susceptible to dephosphorylation with lambda phosphatase. To investigate ENS tau phosphorylation further, primary cultures from rat enteric neurons, which express four isoforms of tau, were pharmacologically manipulated to show that ENS tau phosphorylation state can be regulated, at least in vitro. Our study is the first to characterize tau in the rodent and human ENS. As a whole, our findings provide a basis to unravel the functions of tau in the ENS and to further investigate the possibility of pathological changes in enteric neuropathies and tauopathies.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Enfermedad de Parkinson/patología , Parálisis Supranuclear Progresiva/patología , Proteínas tau/metabolismo , Anciano , Animales , Antiinfecciosos/farmacología , Benzofenantridinas/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Colon/metabolismo , Colon/patología , Embrión de Mamíferos , Sistema Nervioso Entérico/efectos de los fármacos , Femenino , Humanos , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Plexo Mientérico/metabolismo , Embarazo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Plexo Submucoso/metabolismo , Tubulina (Proteína)/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Adulto Joven , Proteínas tau/genética
14.
Acta Neuropathol Commun ; 6(1): 37, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29730992

RESUMEN

In Alzheimer's disease (AD) and related tauopathies, the microtubule-associated protein tau is highly phosphorylated and aggregates to form neurofibrillary tangles that are characteristic of these neurodegenerative diseases. Our previous work has demonstrated that the thousand-and-one amino acid kinases (TAOKs) 1 and 2 phosphorylate tau on more than 40 residues in vitro. Here we show that TAOKs are phosphorylated and active in AD brain sections displaying mild (Braak stage II), intermediate (Braak stage IV) and advanced (Braak stage VI) tau pathology and that active TAOKs co-localise with both pre-tangle and tangle structures. TAOK activity is also enriched in pathological tau containing sarkosyl-insoluble extracts prepared from AD brain. Two new phosphorylated tau residues (T123 and T427) were identified in AD brain, which appear to be targeted specifically by TAOKs. A new small molecule TAOK inhibitor (Compound 43) reduced tau phosphorylation on T123 and T427 and also on additional pathological sites (S262/S356 and S202/T205/S208) in vitro and in cell models. The TAOK inhibitor also decreased tau phosphorylation in differentiated primary cortical neurons without affecting markers of synapse and neuron health. Notably, TAOK activity also co-localised with tangles in post-mortem frontotemporal lobar degeneration (FTLD) brain tissue. Furthermore, the TAOK inhibitor decreased tau phosphorylation in induced pluripotent stem cell derived neurons from FTLD patients, as well as cortical neurons from a transgenic mouse model of tauopathy (Tau35 mice). Our results demonstrate that abnormal TAOK activity is present at pre-tangles and tangles in tauopathies and that TAOK inhibition effectively decreases tau phosphorylation on pathological sites. Thus, TAOKs may represent a novel target to reduce or prevent tau-associated neurodegeneration in tauopathies.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Tauopatías/complicaciones , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Corteza Cerebral , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , L-Lactato Deshidrogenasa/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transfección
15.
Mol Neurobiol ; 55(6): 5125-5136, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28840468

RESUMEN

Dopamine replacement therapy with L-DOPA is the treatment of choice for Parkinson's disease; however, its long-term use is frequently associated with L-DOPA-induced dyskinesia (LID). Many molecules have been implicated in the development of LID, and several of these have been proposed as potential therapeutic targets. However, to date, none of these molecules have demonstrated full clinical efficacy, either because they lie downstream of dopaminergic signaling, or due to adverse side effects. Therefore, discovering new strategies to reduce LID in Parkinson's disease remains a major challenge. Here, we have explored the tyrosine kinase Fyn, as a novel intermediate molecule in the development of LID. Fyn, a member of the Src kinase family, is located in the postsynaptic density, where it regulates phosphorylation of the NR2B subunit of the N-methyl-D-aspartate (NMDA) receptor in response to dopamine D1 receptor stimulation. We have used Fyn knockout and wild-type mice, lesioned with 6-hydroxydopamine and chronically treated with L-DOPA, to investigate the role of Fyn in the induction of LID. We found that mice lacking Fyn displayed reduced LID, ΔFosB accumulation and NR2B phosphorylation compared to wild-type control mice. Pre-administration of saracatinib (AZD0530), an inhibitor of Fyn activity, also significantly reduced LID in dyskinetic wild-type mice. These results support that Fyn has a critical role in the molecular pathways affected during the development of LID and identify Fyn as a novel potential therapeutic target for the management of dyskinesia in Parkinson's disease.


Asunto(s)
Discinesia Inducida por Medicamentos/complicaciones , Discinesia Inducida por Medicamentos/enzimología , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/enzimología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Animales , Benzodioxoles/farmacología , Discinesia Inducida por Medicamentos/patología , Discinesia Inducida por Medicamentos/fisiopatología , Femenino , Levodopa , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Movimiento , Neostriado/metabolismo , Neostriado/patología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Fosforilación , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-fyn/antagonistas & inhibidores , Quinazolinas/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
16.
Neurobiol Aging ; 60: 44-56, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28917666

RESUMEN

The accumulation of cleaved tau fragments in the brain is associated with several tauopathies. For this reason, we recently developed a transgenic mouse that selectively accumulates a C-Terminal 35 kDa human tau fragment (Tau35). These animals develop progressive motor and spatial memory impairment, paralleled by increased hippocampal glycogen synthase kinase 3ß activity. In this neurophysiological study, we focused on the CA1 subfield of the hippocampus, a brain area involved in memory encoding. The accumulation of Tau35 results in a significant increase of short-term facilitation of the synaptic response in the theta frequency range (10 Hz), without affecting basal synaptic transmission and long-term synaptic plasticity. Tau35 expression also alters the intrinsic excitability of CA1 pyramidal neurons. Thus, Tau35 presence is associated with increased and decreased excitability at hyperpolarized and depolarized potentials, respectively. These observations are paralleled by a hyperpolarization of the voltage-sensitivity of noninactivating K+ currents. Further investigation is needed to assess the causal link between such functional alterations and the cognitive and motor impairments previously observed in this model.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Animales , Región CA1 Hipocampal/enzimología , Cognición , Demencia/etiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Trastornos de la Memoria/genética , Ratones Transgénicos , Actividad Motora , Plasticidad Neuronal , Trastornos Psicomotores/genética , Memoria Espacial , Parálisis Supranuclear Progresiva/etiología , Transmisión Sináptica
17.
Sci Rep ; 7(1): 7434, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28785087

RESUMEN

Organotypic brain slice culture models provide an alternative to early stage in vivo studies as an integrated tissue system that can recapitulate key disease features, thereby providing an excellent platform for drug screening. We recently described a novel organotypic 3xTg-AD mouse brain slice culture model with key Alzheimer's disease-like changes. We now highlight the potential of this model for testing disease-modifying agents and show that results obtained following in vivo treatment are replicated in brain slice cultures from 3xTg-AD mice. Moreover, we describe novel effects of the amyloid-binding tetra (ethylene glycol) derivative of benzothiazole aniline, BTA-EG4, on tau. BTA-EG4 significantly reduced tau phosphorylation in the absence of any changes in the amounts of amyloid precursor protein, amyloid-ß or synaptic proteins. The reduction in tau phosphorylation was associated with inactivation of the Alzheimer's disease-relevant major tau kinase, GSK-3. These findings highlight the utility of 3xTg-AD brain slice cultures as a rapid and reliable in vitro method for drug screening prior to in vivo testing. Furthermore, we demonstrate novel tau-directed effects of BTA-EG4 that are likely related to the ability of this agent to inactivate GSK-3. Our findings support the further exploration of BTA-EG4 as a candidate therapeutic for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Polietilenglicoles/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Evaluación Preclínica de Medicamentos , Humanos , Cloruro de Litio/farmacología , Ratones , Ratones Transgénicos , Modelos Biológicos , Oligopéptidos/farmacología , Técnicas de Cultivo de Órganos/métodos , Fosforilación/efectos de los fármacos , Polietilenglicoles/química
18.
Acta Neuropathol ; 133(5): 665-704, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28386764

RESUMEN

Tau is well established as a microtubule-associated protein in neurons. However, under pathological conditions, aberrant assembly of tau into insoluble aggregates is accompanied by synaptic dysfunction and neural cell death in a range of neurodegenerative disorders, collectively referred to as tauopathies. Recent advances in our understanding of the multiple functions and different locations of tau inside and outside neurons have revealed novel insights into its importance in a diverse range of molecular pathways including cell signalling, synaptic plasticity, and regulation of genomic stability. The present review describes the physiological and pathophysiological properties of tau and how these relate to its distribution and functions in neurons. We highlight the post-translational modifications of tau, which are pivotal in defining and modulating tau localisation and its roles in health and disease. We include discussion of other pathologically relevant changes in tau, including mutation and aggregation, and how these aspects impinge on the propensity of tau to propagate, and potentially drive neuronal loss, in diseased brain. Finally, we describe the cascade of pathological events that may be driven by tau dysfunction, including impaired axonal transport, alterations in synapse and mitochondrial function, activation of the unfolded protein response and defective protein degradation. It is important to fully understand the range of neuronal functions attributed to tau, since this will provide vital information on its involvement in the development and pathogenesis of disease. Such knowledge will enable determination of which critical molecular pathways should be targeted by potential therapeutic agents developed for the treatment of tauopathies.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/patología , Humanos , Enfermedades Neurodegenerativas/patología , Procesamiento Proteico-Postraduccional/fisiología , Tauopatías/patología
19.
Acta Neuropathol ; 134(1): 129-149, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28337542

RESUMEN

α-Synuclein is strongly linked to Parkinson's disease but the molecular targets for its toxicity are not fully clear. However, many neuronal functions damaged in Parkinson's disease are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling involves close physical associations between the two organelles that are mediated by binding of the integral ER protein vesicle-associated membrane protein-associated protein B (VAPB) to the outer mitochondrial membrane protein, protein tyrosine phosphatase-interacting protein 51 (PTPIP51). VAPB and PTPIP51 thus act as a scaffold to tether the two organelles. Here we show that α-synuclein binds to VAPB and that overexpression of wild-type and familial Parkinson's disease mutant α-synuclein disrupt the VAPB-PTPIP51 tethers to loosen ER-mitochondria associations. This disruption to the VAPB-PTPIP51 tethers is also seen in neurons derived from induced pluripotent stem cells from familial Parkinson's disease patients harbouring pathogenic triplication of the α-synuclein gene. We also show that the α-synuclein induced loosening of ER-mitochondria contacts is accompanied by disruption to Ca2+ exchange between the two organelles and mitochondrial ATP production. Such disruptions are likely to be particularly damaging to neurons that are heavily dependent on correct Ca2+ signaling and ATP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Proteínas de Transporte Vesicular/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cationes Bivalentes/metabolismo , Línea Celular Tumoral , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas Tirosina Fosfatasas/metabolismo , Ratas Sprague-Dawley , alfa-Sinucleína/genética
20.
Cell Death Dis ; 8(3): e2671, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300838

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Membrana Celular/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Emparejamiento Cromosómico/fisiología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos/metabolismo , Neuronas/metabolismo , Fosforilación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...