Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros










Intervalo de año de publicación
1.
Transl Neurodegener ; 11(1): 34, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35773715

RESUMEN

BACKGROUND: Neuronal dysfunction and degeneration linked to α-synuclein (αS) pathology is thought to be responsible for the progressive nature of Parkinson's disease and related dementia with Lewy bodies. Studies have indicated bidirectional pathological relationships between αS pathology and tau abnormalities. We recently showed that A53T mutant human αS (HuαS) can cause post-synaptic and cognitive deficits that require microtubule-associated protein tau expression. However, the role of tau in the development of αS pathology and subsequent neuronal dysfunction has been controversial. Herein, we set out to determine the role of tau in the onset and progression of αS pathology (α-synucleinopathy) using a transgenic mouse model of α-synucleinopathy lacking mouse tau expression. METHODS: Transgenic mice expressing A53T mutant HuαS (TgA53T) were crossed with mTau-/- mice to generate TgA53T/mTau-/-. To achieve more uniform induction of α-synucleinopathy in mice, we used intramuscular injections of αS preformed fibrils (PFF) in non-transgenic (nTg), TgA53T, TgA53T/mTau-/-, and mTau-/- mice. Motor behavior was analyzed at 70 days post inoculation (dpi) of PFF and tissues for biochemical and neuropathological analysis were collected at 40 dpi, 70 dpi, and end stage. RESULTS: Loss of tau expression significantly delayed the onset of motor deficits in the TgA53T model and the progression of α-synucleinopathy disease, as evidenced by a significant reduction in histopathological and behavioral markers of neurodegeneration and disease, and a significant improvement in survival. In vitro application of PFF to primary mouse hippocampal neurons demonstrated no changes in PFF uptake and processing or pS129 αS aggregation as a function of tau expression. However, PFF-induced neurotoxicity, including morphological deficits in nTg neurons, was prevented with tau removal. CONCLUSIONS: Collectively, our data suggest that tau is likely acting downstream of αS pathology to affect neuronal homeostasis and survival. This work further supports the investigation of tau in α-synucleinopathies to identify novel disease-modifying therapeutic strategies.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Proteínas tau , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sinucleinopatías/genética , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Proteínas tau/biosíntesis , Proteínas tau/deficiencia , Proteínas tau/genética
2.
Cell Rep ; 37(3): 109855, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686344

RESUMEN

The protein tau has been implicated in many brain disorders. In animal models, tau reduction suppresses epileptogenesis of diverse causes and ameliorates synaptic and behavioral abnormalities in various conditions associated with excessive excitation-inhibition (E/I) ratios. However, the underlying mechanisms are unknown. Global genetic ablation of tau in mice reduces the action potential (AP) firing and E/I ratio of pyramidal cells in acute cortical slices without affecting the excitability of these cells. Tau ablation reduces the excitatory inputs to inhibitory neurons, increases the excitability of these cells, and structurally alters their axon initial segments (AISs). In primary neuronal cultures subjected to prolonged overstimulation, tau ablation diminishes the homeostatic response of AISs in inhibitory neurons, promotes inhibition, and suppresses hypersynchrony. Together, these differential alterations in excitatory and inhibitory neurons help explain how tau reduction prevents network hypersynchrony and counteracts brain disorders causing abnormally increased E/I ratios.


Asunto(s)
Potenciales Postsinápticos Excitadores , Potenciales Postsinápticos Inhibidores , Interneuronas/metabolismo , Inhibición Neural , Vías Nerviosas/metabolismo , Células Piramidales/metabolismo , Corteza Somatosensorial/metabolismo , Proteínas tau/deficiencia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Células Cultivadas , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/fisiopatología , Femenino , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/citología , Plasticidad Neuronal , Corteza Somatosensorial/citología , Factores de Tiempo , Proteínas tau/genética
3.
Genes (Basel) ; 12(6)2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34198629

RESUMEN

Traumatic brain injuries, a leading cause of death and disability worldwide, are caused by a severe impact to the head that impairs physiological and psychological function. In addition to severity, type and brain area affected, brain injury outcome is also influenced by the biological sex of the patient. Traumatic brain injury triggers accumulation of Tau protein and the subsequent development of Tauopathies, including Alzheimer's disease and Chronic traumatic encephalopathy. Recent studies report differences in Tau network connections between healthy males and females, but the possible role of Tau in sex-dependent outcome to brain injury is unclear. Thus, we aimed to determine if Tau ablation would alleviate sex dependent outcomes in injured flies. We first assessed motor function and survival in tau knock-out flies and observed sex-differences in climbing ability, but no change in locomotor activity in either sex post-injury. Sex differences in survival time were also observed in injured tau deficient flies with a dramatically higher percent of female death within 24 h than males. Additionally, 3'mRNA-Seq studies in isolated fly brains found that tau deficient males show more gene transcript changes than females post-injury. Our results suggest that sex differences in TBI outcome and recovery are not dependent on the presence of Tau in Drosophila.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Proteínas de Drosophila/genética , Proteínas tau/genética , Animales , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/genética , Proteínas de Drosophila/deficiencia , Drosophila melanogaster , Femenino , Masculino , Movimiento , Factores Sexuales , Transcriptoma , Proteínas tau/deficiencia
4.
Int J Neurosci ; 131(6): 527-535, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32408779

RESUMEN

BACKGROUND: Humanin (HN) is an extensive neuroprotective peptide. This study aims to investigate the neuroprotective effects of HN on Calyculin A (CA)-induced neurotoxicities in cortical neurons and the underlying mechanism. METHODS: CA was added into the cultured cortical neurons to induce neurotoxicity. Cortical neurons were preincubated with HN which plays a protective role. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH), and Calcein-AM were applied to evaluate the neural insults. Caspase 3 signal and Tunnel were performed to test neural apoptosis. Western blot analysis was used to detect the expressions of phosphorylated tau. The corresponding kits were used to measure the contents of malondialdehyde (MDA) and superoxide dismutase (SOD), and the activity of PP2A, respectively. RESULTS: HN preincubation preserved cell viability, protected the neurons, alleviated oxidative stress, and reserved PP2A activity. It also blocked tau overphosphorylation at Ser199/202, Ser396, and Thr231 sites and protected neurons against CA-induced insults. CONCLUSION: These results suggest that HN may serve as a potential therapeutic agent to prevent the pathological changes induced by CA via modulating the activity of PP2A and oxidative stress in neurodegenerative diseases.


Asunto(s)
Carcinógenos/toxicidad , Corteza Cerebral/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Toxinas Marinas/toxicidad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oxazoles/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/efectos de los fármacos , Proteínas tau/deficiencia , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Ratas
5.
Cells ; 9(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947657

RESUMEN

Tau dysfunction is common in several neurodegenerative diseases including Alzheimer's disease (AD) and frontotemporal dementia (FTD). Affective symptoms have often been associated with aberrant tau pathology and are commonly comorbid in patients with tauopathies, indicating a connection between tau functioning and mechanisms of depression. The current study investigated depression-like behavior in Mapt-/- mice, which contain a targeted deletion of the gene coding for tau. We show that 6-month Mapt-/- mice are resistant to depressive behaviors, as evidenced by decreased immobility time in the forced swim and tail suspension tests, as well as increased escape behavior in a learned helplessness task. Since depression has also been linked to deficient adult neurogenesis, we measured neurogenesis in the hippocampal dentate gyrus and subventricular zone using 5-bromo-2-deoxyuridine (BrdU) labeling. We found that neurogenesis is increased in the dentate gyrus of 14-month-old Mapt-/- brains compared to wild type, providing a potential mechanism for their behavioral phenotypes. In addition to the hippocampus, an upregulation of proteins involved in neurogenesis was observed in the frontal cortex and amygdala of the Mapt-/- mice using proteomic mass spectrometry. All together, these findings suggest that tau may have a role in the depressive symptoms observed in many neurodegenerative diseases and identify tau as a potential molecular target for treating depression.


Asunto(s)
Hipocampo/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas tau/deficiencia , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología
7.
Sci Rep ; 9(1): 9116, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31235881

RESUMEN

Tau is a microtubule-associated neuronal protein found mainly in axons. However, increasing evidence indicates that it is also present in dendrites, where it serves as an essential mediator of synaptic NMDA (N-methyl-D-aspartate) receptor-dependent excitotoxicity. Of note, NMDA receptors can also be found outside synapses in the plasma membrane, and activation of extrasynaptic NMDA receptors has been shown to be more linked to excitotoxicity than the activation of synaptic ones. Little is known about the role of Tau in the activity of extrasynaptic NMDA receptors. Thus, we have used a Tau knockout mouse model (Tau-/- mice) to analyze the consequences of Tau absence in extrasynaptic NMDA receptor activity. We demonstrate that absence of Tau leads to a decrease in functional extrasynaptic NMDA receptors in the hippocampus. We propose that this impairment in extrasynaptic NMDA receptor activity may contribute to the well-known neuroprotective effect associated with Tau deficiency under pathological conditions.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Proteínas tau/metabolismo , Animales , Fenómenos Electrofisiológicos , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/fisiología , Ratones , Neuronas/metabolismo , Proteínas tau/deficiencia
8.
Acta Neuropathol Commun ; 7(1): 26, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808415

RESUMEN

Cerebral amyloid angiopathy (CAA) is typified by the cerebrovascular deposition of amyloid. Currently, there is no clear understanding of the mechanisms underlying the contribution of CAA to neurodegeneration. Despite the fact that CAA is highly associated with accumulation of Aß, other types of amyloids have been shown to associate with the vasculature. Interestingly, in many cases, vascular amyloidosis is accompanied by significant tau pathology. However, the contribution of tau to neurodegeneration associated to CAA remains to be determined. We used a mouse model of Familial Danish Dementia (FDD), a neurodegenerative disease characterized by the accumulation of Danish amyloid (ADan) in the vasculature, to characterize the contribution of tau to neurodegeneration associated to CAA. We performed histological and biochemical assays to establish tau modifications associated with CAA in conjunction with cell-based and electrophysiological assays to determine the role of tau in the synaptic dysfunction associated with ADan. We demonstrated that ADan aggregates induced hyperphosphorylation and misfolding of tau. Moreover, in a mouse model for CAA, we observed tau oligomers closely associated to astrocytes in the vicinity of vascular amyloid deposits. We finally determined that the absence of tau prevents synaptic dysfunction induced by ADan oligomers. In addition to demonstrating the effect of ADan amyloid on tau misfolding, our results provide compelling evidence of the role of tau in neurodegeneration associated with ADan-CAA and suggest that decreasing tau levels could be a feasible approach for the treatment of CAA.


Asunto(s)
Angiopatía Amiloide Cerebral/genética , Angiopatía Amiloide Cerebral/metabolismo , Proteínas tau/deficiencia , Proteínas tau/genética , Secuencia de Aminoácidos , Amiloidosis/genética , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Catarata/genética , Catarata/metabolismo , Catarata/patología , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/metabolismo , Ataxia Cerebelosa/patología , Angiopatía Amiloide Cerebral/patología , Sordera/genética , Sordera/metabolismo , Sordera/patología , Demencia/genética , Demencia/metabolismo , Demencia/patología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
9.
Acta Neuropathol Commun ; 6(1): 57, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29976255

RESUMEN

Parkinson's disease is diagnosed upon the presentation of motor symptoms, resulting from substantial degeneration of dopaminergic neurons in the midbrain. Prior to diagnosis, there is a lengthy prodromal stage in which non-motor symptoms, including olfactory deficits (hyposmia), develop. There is limited information about non-motor impairments and there is a need for directed research into these early pathogenic cellular pathways that precede extensive dopaminergic death in the midbrain. The protein tau has been identified as a genetic risk factor in the development of sporadic PD. Tau knockout mice have been reported as an age-dependent model of PD, and this study has demonstrated that they develop motor deficits at 15-months-old. We have shown that at 7-month-old tau knockout mice present with an overt hyposmic phenotype. This olfactory deficit correlates with an accumulation of α-synuclein, as well as autophagic impairment, in the olfactory bulb. This pathological feature becomes apparent in the striatum and substantia nigra of 15-month-old tau knockout mice, suggesting the potential for a spread of disease. Initial primary cell culture experiments have demonstrated that ablation of tau results in the release of α-synuclein enriched exosomes, providing a potential mechanism for disease spread. These alterations in α-synuclein level as well as a marked autophagy impairment in the tau knockout primary cells recapitulate results seen in the animal model. These data implicate a pathological role for tau in early Parkinson's disease.


Asunto(s)
Trastornos del Olfato/etiología , Trastornos del Olfato/genética , Enfermedad de Parkinson/complicaciones , Proteínas tau/deficiencia , Factores de Edad , Animales , Autofagia , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Exosomas/metabolismo , Exosomas/patología , Exosomas/ultraestructura , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Odorantes , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/patología , Enfermedad de Parkinson/patología , Desempeño Psicomotor/fisiología , Proteína Sequestosoma-1/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/genética
10.
J Alzheimers Dis ; 65(1): 265-281, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30040731

RESUMEN

The tau spreading hypothesis provides rationale for passive immunization with an anti-tau monoclonal antibody to block seeding by extracellular tau aggregates as a disease-modifying strategy for the treatment of Alzheimer's disease (AD) and potentially other tauopathies. As the biochemical and biophysical properties of the tau species responsible for the spatio-temporal sequences of seeding events are poorly defined, it is not yet clear which epitope is preferred for obtaining optimal therapeutic efficacy. Our internal tau antibody collection has been generated by immunizations with different tau species: aggregated- and non-aggregated tau and human postmortem AD brain-derived tau fibrils. In this communication, we describe and characterize a set of these anti-tau antibodies for their biochemical and biophysical properties, including binding, tissue staining by immunohistochemistry, and epitope. The antibodies bound to different domains of the tau protein and some were demonstrated to be isoform-selective (PT18 and hTau56) or phospho-selective (PT84). Evaluation of the antibodies in cellular- and in vivo seeding assays revealed clear differences in maximal efficacy. Limited proteolysis experiments support the hypothesis that some epitopes are more exposed than others in the tau seeds. Moreover, antibody efficacy seems to depend on the structural properties of fibrils purified from tau Tg mice- and postmortem human AD brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Anticuerpos Monoclonales/metabolismo , Encéfalo/metabolismo , Proteínas tau/inmunología , Animales , Mapeo Epitopo , Femenino , Células HEK293 , Humanos , Inmunización Pasiva , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Resonancia por Plasmón de Superficie , Proteínas tau/deficiencia , Proteínas tau/genética
11.
Aging Cell ; 17(4): e12775, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29749079

RESUMEN

Misfolded and hyperphosphorylated tau accumulates in several neurodegenerative disorders including Alzheimer's disease, frontotemporal dementia with Parkinsonism, corticobasal degeneration, progressive supranuclear palsy, Down syndrome, and Pick's disease. Tau is a microtubule-binding protein, and its role in microtubule stabilization is well defined. In contrast, while growing evidence suggests that tau is also involved in synaptic physiology, a complete assessment of tau function in the adult brain has been hampered by robust developmental compensation of other microtubule-binding proteins in tau knockout mice. To circumvent these developmental compensations and assess the role of tau in the adult brain, we generated an adeno-associated virus (AAV) expressing a doxycycline-inducible short-hairpin (Sh) RNA targeted to tau, herein referred to as AAV-ShRNATau. We performed bilateral stereotaxic injections in 7-month-old C57Bl6/SJL wild-type mice with either the AAV-ShRNATau or a control AAV. We found that acute knockdown of tau in the adult hippocampus significantly impaired motor coordination and spatial memory. Blocking the expression of the AAV-ShRNATau, thereby allowing tau levels to return to control levels, restored motor coordination and spatial memory. Mechanistically, the reduced tau levels were associated with lower BDNF levels, reduced levels of synaptic proteins associated with learning, and decreased spine density. We provide compelling evidence that tau is necessary for motor and cognitive function in the adult brain, thereby firmly supporting that tau loss-of-function may contribute to the clinical manifestations of many tauopathies. These findings have profound clinical implications given that anti-tau therapies are in clinical trials for Alzheimer's disease.


Asunto(s)
Hipocampo/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Trastornos de la Memoria/metabolismo , Proteínas tau/deficiencia , Proteínas tau/metabolismo , Animales , Dependovirus/aislamiento & purificación , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas tau/aislamiento & purificación
12.
Neuroscience ; 373: 52-59, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337233

RESUMEN

Tau protein participates in microtubule stabilization, axonal transport, and protein trafficking. Loss of normal tau function will exert a negative effect. However, current knowledge on the impact of tau deficiency on the motor behavior and related neurobiological changes is controversial. In this study, we examined motor functions and analyzed several proteins implicated in the maintenance of midbrain dopaminergic (DA) neurons (mDANs) function of adult and aged tau+/+, tau+/-, tau-/- mice. We found tau deficiency could not induce significant motor disorders. However, we discovered lower expression levels of transcription factors Orthodenticle homeobox 2 (OTX2) of mDANs in older aged mice. Compared with age-matched tau+/+ mice, there were 54.1% lower (p = 0.0192) OTX2 protein (OTX2-fluorescence intensity) in VTA DA neurons of tau+/- mice and 43.6% lower (p = 0.0249) OTX2 protein in VTA DA neurons of tau-/- mice at 18 months old. Combined with the relevant reports, our results suggested that tau deficiency alone might not be enough to mimic the pathology of Parkinson's disease. However, OTX2 down-regulation indicates that mDANs of tau-deficient mice will be more sensitive to toxic damage from MPTP.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Actividad Motora/fisiología , Factores de Transcripción Otx/metabolismo , Área Tegmental Ventral/metabolismo , Proteínas tau/deficiencia , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/patología , Regulación hacia Abajo/fisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fuerza Muscular/fisiología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Área Tegmental Ventral/patología , Proteínas tau/genética
13.
Nat Commun ; 8(1): 473, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28883427

RESUMEN

Neuronal excitotoxicity induced by aberrant excitation of glutamatergic receptors contributes to brain damage in stroke. Here we show that tau-deficient (tau-/-) mice are profoundly protected from excitotoxic brain damage and neurological deficits following experimental stroke, using a middle cerebral artery occlusion with reperfusion model. Mechanistically, we show that this protection is due to site-specific inhibition of glutamate-induced and Ras/ERK-mediated toxicity by accumulation of Ras-inhibiting SynGAP1, which resides in a post-synaptic complex with tau. Accordingly, reducing SynGAP1 levels in tau-/- mice abolished the protection from pharmacologically induced excitotoxicity and middle cerebral artery occlusion-induced brain damage. Conversely, over-expression of SynGAP1 prevented excitotoxic ERK activation in wild-type neurons. Our findings suggest that tau mediates excitotoxic Ras/ERK signaling by controlling post-synaptic compartmentalization of SynGAP1.Excitotoxicity contributes to neuronal injury following stroke. Here the authors show that tau promotes excitotoxicity by a post-synaptic mechanism, involving site-specific control of ERK activation, in a mouse model of stroke.


Asunto(s)
Lesiones Encefálicas/genética , Modelos Animales de Enfermedad , Accidente Cerebrovascular/genética , Proteínas tau/genética , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Transducción de Señal/genética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Sinaptosomas/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas tau/deficiencia
14.
Prog Neuropsychopharmacol Biol Psychiatry ; 79(Pt B): 452-461, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28779908

RESUMEN

BACKGROUND: Tau is a microtubule-associated protein that becomes pathological when it undergoes hyperphosphorylation and aggregation as seen in Alzheimer's disease (AD). AD is mostly sporadic, with environmental, biological and/or genetic risks factors, interacting together to promote the disease. In the past decade, reports have suggested that obesity in midlife could be one of these risk factors. On the other hand, caloric restriction and physical exercise have been reported to reduce the incidence and outcome of obesity as well as AD. METHODS: We evaluated the impact of voluntary physical exercise and caloric restriction on tau pathology during 2months in hTau mice under high caloric diet in order to evaluate if these strategies could prevent AD-like pathology in obese conditions. RESULTS: We found no effects of obesity induced by Western diet on both Tau phosphorylation and aggregation compared to controls. However, exercise reduced tau phosphorylation while caloric restriction exacerbated its aggregation in the brains of obese hTau mice. We then examined the mechanisms underlying changes in tau phosphorylation and aggregation by exploring major tau kinases and phosphatases and key proteins involved in autophagy. However, there were no significant effects of voluntary exercise and caloric restriction on these proteins in hTau mice that could explain our results. CONCLUSION: In this study, we report differential effects of voluntary treadmill exercise and caloric restriction on tau pathogenesis in our obese mice, namely beneficial effect of exercise on tau phosphorylation and deleterious effect of caloric restriction on tau aggregation. Our results suggest that lifestyle strategies used to reduce metabolic disorders and AD must be selected and studied carefully to avoid exacerbation of pathologies.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Restricción Calórica/efectos adversos , Dieta Occidental/efectos adversos , Actividad Motora/fisiología , Obesidad/fisiopatología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/patología , Obesidad/terapia , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/fisiopatología , Agregación Patológica de Proteínas/terapia , Volición , Proteínas tau/deficiencia , Proteínas tau/genética
15.
Acta Neuropathol Commun ; 5(1): 58, 2017 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-28760159

RESUMEN

Tauopathies are a group of neurodegenerative disorders, including Alzheimer's disease, defined by the presence of brain pathological inclusions comprised of abnormally aggregated and highly phosphorylated tau protein. The abundance of brain tau aggregates correlates with disease severity and select phospho-tau epitopes increase at early stages of disease. We generated and characterized a series of novel monoclonal antibodies directed to tau phosphorylated at several of these phospho-epitopes, including Ser396/Ser404, Ser404 and Thr205. We also generated phosphorylation independent antibodies against amino acid residues 193-211. We show that most of these antibodies are highly specific for tau and strongly recognize pathological inclusions in human brains and in a transgenic mouse model of tauopathy. They also reveal epitope-specific differences in the biochemical properties of Alzheimer's disease sarkosyl-insoluble tau. These new reagents will be useful for investigating the progression of tau pathology and further as tools to target the cellular transmission of tau pathology.


Asunto(s)
Anticuerpos Monoclonales , Epítopos , Proteínas tau/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Hibridomas/metabolismo , Inmunohistoquímica , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutación , Fosforilación , Proteínas Recombinantes/metabolismo , Proteínas tau/deficiencia , Proteínas tau/genética
16.
Neurosci Lett ; 653: 276-282, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28554859

RESUMEN

Microtubule-associated protein tau has crucial roles not only in the formation of some neurodegenerative disorders but also in normal synaptic functions, although its contributions to these are still unclear. Here, to reveal the influence of tau deletion on trafficking of synaptic receptors, we investigated the distribution of GluA2-containing AMPA-type glutamate receptors (AMPARs) within neuronal dendrites in wild-type and tau-deficient neurons using biochemical and laser-confocal imaging techniques. Under basal conditions, expression of GluA2 at tau-deficient synapses was almost normal; however, its level within dendrites in tau-deficient neurons was greater than that in wild-type neurons. After NMDA treatment, a decrease in GluA2-containing AMPARs at synapses was observed in wild-type neurons, but not in tau-deficient neurons. Single-cell imaging of GluA2 within dendrites demonstrated that wild-type neurons, but not tau-deficient neurons, showed enlargement of GluA2 puncta. Interestingly, we also found that NMDA rapidly reduced the number of GluA2 puncta without changing their size in tau-deficient neurons but not wild-type neurons. These results demonstrate the multiple contributions of tau to the maintenance of dynamic AMPAR trafficking within dendrites during both stimulated and unstimulated conditions.


Asunto(s)
Dendritas/metabolismo , Receptores AMPA/metabolismo , Proteínas tau/metabolismo , Animales , Femenino , Masculino , Ratones , Transporte de Proteínas , Proteínas tau/deficiencia
17.
J Alzheimers Dis ; 56(2): 849-860, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28059795

RESUMEN

Aggregation of tau protein into intracellular deposits is a pathognomonic feature of tauopathies such as Alzheimer's disease (AD) and lowering tau is a prominent therapeutic strategy under development. However, the physiological function of tau protein is not well known, particularly in the periphery. Lowering tau protein risks disrupting its physiological role leading to unwanted effects. In this study, the presence of tau protein in cardiac tissue is confirmed and the functional role in the cardiovascular system and the consequences of its loss were explored. Isolated right and left atria and small mesenteric arteries from wild type and tau deficient (KO) mice of two age groups (13 and 23 months old) were used to assess cardiovascular phenotypes. Tau KO mice showed an increased systolic blood pressure and cardiac hypertrophy at 13 months, which was accompanied by a significantly lower right atrial rate and a subtle decrease in the maximum contractility to calcium, isoprenaline, and electrical sympathetic nerve stimulation. Aging tau KO mice to 23 months resulted in cardiac hypertrophy with significantly attenuated left atrial contractility, increased blood pressure, and sensitivity of isolated mesenteric arteries to angiotensin II contraction and isoprenaline relaxation compared to their younger counterparts. This study supports a functional role of tau in the heart and loss of this protein leads to a deterioration in cardiovascular performance which worsens with age. Taken together, these results provide insight into the peripheral function of tau protein, and give caution to the therapeutic strategy of lowering tau protein.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/metabolismo , Proteínas tau/deficiencia , Envejecimiento/patología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Enfermedades Cardiovasculares/patología , Femenino , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Hipertrofia/metabolismo , Hipertrofia/patología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Técnicas de Cultivo de Tejidos , Proteínas tau/genética
18.
Neuroscience ; 329: 98-111, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27167086

RESUMEN

htau mice are deficient of murine tau but express all six human tau isoforms, leading to gradual tau misprocessing and aggregation in brain areas relevant to Alzheimer's disease. While histopathological changes in htau mice have been researched in the past, we focused here on functional consequences of human tau accumulation. htau mice and their background controls - murine tau knock-out (mtau(-/-)) and C57Bl/6J mice - underwent a comprehensive trial battery to investigate species-specific behavior, locomotor activity, emotional responses, exploratory traits, spatial and recognition memory as well as acquisition, retention and extinction of contextual fear at two, four, six, nine and twelve months of age. In htau mice, tau pathology was already present at two months of age, whereas deficits in food burrowing and spatial working memory were first noted at four months of age. At later stages the presence of human tau on a mtau(-/-) background appeared to guard cognitive performance; as mtau(-/-) but not htau mice differed from C57Bl/6J mice in the food burrowing, spontaneous alternation and object discrimination tasks. Aging mtau(-/-) mice also exhibited increased body mass and locomotor activity. These data highlight that reduced food-burrowing performance was the most robust aspect of the htau phenotype with aging. htau and mtau(-/-) deficits in food burrowing pointed at the necessity of intact tau systems for daily life activities. While some htau and mtau(-/-) deficits overlap, age differences between the two genotypes may reflect distinct functional effects and compared to C57Bl/6J mice, the htau phenotype appeared stronger than the mtau(-/-) phenotype at young ages but milder with aging.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/psicología , Conducta Animal/fisiología , Actividad Motora/fisiología , Proteínas tau/deficiencia , Proteínas tau/metabolismo , Animales , Corteza Cerebral/metabolismo , Cognición/fisiología , Humanos , Aprendizaje/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Isoformas de Proteínas , Especificidad de la Especie , Proteínas tau/genética
19.
PLoS One ; 11(4): e0153724, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27070146

RESUMEN

Alzheimer's disease (AD) researchers have struggled for decades to draw a causal link between extracellular Aß aggregation and intraneuronal accumulation of microtubule-associated protein tau. The amyloid cascade hypothesis posits that Aß deposition promotes tau hyperphosphorylation, tangle formation, cell loss, vascular damage, and dementia. While the genetics of familial AD and the pathological staging of sporadic disease support this sequence of events, attempts to examine the molecular mechanism in transgenic animal models have largely relied on models of other inherited tauopathies as the basis for testing the interaction with Aß. In an effort to more accurately model the relationship between Aß and wild-type tau in AD, we intercrossed mice that overproduce human Aß with a tau substitution model in which all 6 isoforms of the human protein are expressed in animals lacking murine tau. We selected an amyloid model in which pathology was biased towards the entorhinal region so that we could further examine whether the anticipated changes in tau phosphorylation occurred at the site of Aß deposition or in synaptically connected regions. We found that Aß and tau had independent effects on locomotion, learning, and memory, but found no behavioral evidence for an interaction between the two transgenes. Moreover, we saw no indication of amyloid-induced changes in the phosphorylation or aggregation of human tau either within the entorhinal area or elsewhere. These findings suggest that robust amyloid pathology within the medial temporal lobe has little effect on the metabolism of wild type human tau in this model.


Asunto(s)
Amiloide/metabolismo , Conducta Animal , Proteínas tau/genética , Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Condicionamiento Psicológico , Miedo/psicología , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Fosforilación , Proteínas tau/deficiencia , Proteínas tau/metabolismo
20.
Acta Neuropathol Commun ; 4: 32, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27037086

RESUMEN

In Alzheimer's disease (AD), the distribution and density of neurofibrillary tangles, a histological hallmark comprised predominately of phosphorylated tau protein, follows a distinct pattern through anatomically connected brain regions. Studies in transgenic mice engineered to regionally confine tau expression have suggested spreading of tau within neural networks. Furthermore, injection of protein lysates isolated from brains of transgenic mice or patients with tauopathies, including AD, were shown to behave like seeds, accelerating tau pathology and tangle formation in predisposed mice. However, it remains unclear how the initiation of primary aggregation events occurs and what triggers further dissemination throughout the neural system. To consolidate these findings, we pursued an alternative approach to assess the spreading of endogenous phosphorylated tau. To generate endogenous seeds, 130 nl of 100 µM protein phosphatase 2A inhibitor okadaic acid (OA) was injected unilaterally into the amygdala of 8-month-old C57Bl/6 wild-type mice. OA was detected in brain tissue by ELISA, and found to be restricted to the injected hemispheric quadrant, where it remained detectable a week post-injection. OA injection induced tau phosphorylation that was observed not only at the injection site but also in anatomically distinct areas across both hemispheres, including the cortex and hippocampus 24 h post-injection. An increase in insoluble tau was also observed in both hemispheres of injected brains by 7 days. Furthermore, thioflavin-S detected protein aggregation at the injection site and in the cortex of both injected and contralateral hemispheres. OA injection induced no thioflavin-positivity in tau knock-out mice. The data demonstrates that a local OA insult can rapidly initiate changes in protein phosphorylation, solubility and aggregation at anatomically distant sites. This model suggests that tau phosphorylation can be both a primary response to an insult, and a secondary response communicated to non-exposed brains regions. The study highlights the use of OA to assist in understanding the initiation of tau spreading in vivo.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácido Ocadaico/farmacología , Agregación Patológica de Proteínas/inducido químicamente , Proteínas tau/metabolismo , Animales , Encéfalo/anatomía & histología , Inhibidores Enzimáticos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Ocadaico/metabolismo , Fosforilación/efectos de los fármacos , Factores de Tiempo , Proteínas tau/deficiencia , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...