Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Curr Issues Mol Biol ; 43(1): 405-422, 2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-34205542

RESUMEN

This study confirmed the ameliorating effect of immature persimmon (Diospyros kaki) ethanolic extract (IPEE) on neuronal cytotoxicity in amyloid beta (Aß)1-42-induced ICR mice. The administration of IPEE ameliorated the cognitive dysfunction in Aß1-42-induced mice by improving the spatial working memory, the short-term and long-term memory functions. IPEE protected the cerebral cholinergic system, such as the acetylcholine (ACh) level and acetylcholinesterase (AChE) activity, and antioxidant system, such as the superoxide dismutase (SOD), reduced glutathione (GSH) and malondialdehyde (MDA) contents. In addition, mitochondrial dysfunction against Aß1-42-induced toxicity was reduced by regulating the reactive oxygen species (ROS), mitochondrial membrane potential and ATP contents. In addition, IPEE regulated the expression levels of tau signaling, such as TNF-α, p-JNK, p-Akt, p-GSK3ß, p-tau, p-NF-κB, BAX and caspase 3. Finally, gallic acid, ellagic acid and quercetin 3-O-(6″-acetyl-glucoside) were identified as the physiological compounds of IPEE using ultra-performance liquid chromatography ion mobility separation quadrupole time-of-flight/tandem mass spectrometry (UPLC IMS Q-TOF/MS2).


Asunto(s)
Disfunción Cognitiva/prevención & control , Diospyros/química , Frutas/química , Extractos Vegetales/farmacología , Tauopatías/prevención & control , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Péptidos beta-Amiloides , Animales , Antioxidantes/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Etanol/química , Aprendizaje por Laberinto/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones Endogámicos ICR , Fragmentos de Péptidos , Extractos Vegetales/química , Especies Reactivas de Oxígeno/metabolismo , Tauopatías/inducido químicamente , Tauopatías/metabolismo , Proteínas tau/metabolismo
2.
Neurochem Int ; 148: 105082, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34052296

RESUMEN

Since the role of estrogen in postmenauposal-associated dementia is still debatable, this issue urges the search for other medications. Dimethyl fumarate (DMF) is a drug used for the treatment of multiple sclerosis and has shown a neuroprotective effect against other neurodegenerative diseases. Accordingly, the present study aimed to evaluate the effect of DMF on an experimental model of Alzheimer disease (AD) using D-galactose (D-Gal) administered to ovariectomized (OVX) rats, resembling a postmenopausal dementia paradigm. Adult 18-month old female Wistar rats were allocated into sham-operated and OVX/D-Gal groups that were either left untreated or treated with DMF for 56 days starting three weeks after sham-operation or ovariectomy. DMF succeeded to ameliorate cognitive (learning/short- and long-term memory) deficits and to enhance the dampened overall activity (NOR, Barnes-/Y-maze tests). These behavioral upturns were associated with increased intact neurons (Nissl stain) and a reduction in OVX/D-Gal-mediated hippocampal CA1 neurodegeneration and astrocyte activation assessed as GFAP immunoreactivity. Mechanistically, DMF suppressed the hippocampal contents of AD-surrogate markers; viz., apolipoprotein (APO)-E1, BACE1, Aß42, and hyperphosphorylated Tau. Additionally, DMF has augmented the neuroprotective parameters p-AKT, its downstream target CREB and BDNF. Besides, it activated AMPK, and enhanced SIRT-1, as well as antioxidant defenses (SOD, GSH). On the other hand, DMF inhibited the transcription factor NF-κB, IL-1ß, adiponectin/adiponectin receptor type (AdipoR)1, GSK-3ß, and MDA. Accordingly, in this postmenopausal AD model, DMF treatment by pursuing the adiponectin/AdipoR1, AMPK/SIRT-1, AKT/CREB/BDNF, AKT/GSK-3ß, and APO-E1 quartet hampered the associated tauo-/amyloidopathy and NF-κB-mediated oxidative/inflammatory responses to advance insights into its anti-amnesic effect.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Amiloidosis/tratamiento farmacológico , Dimetilfumarato/farmacología , Fármacos Neuroprotectores/farmacología , Ovariectomía , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tauopatías/tratamiento farmacológico , Adiponectina/genética , Enfermedad de Alzheimer/inducido químicamente , Amiloidosis/inducido químicamente , Amiloidosis/psicología , Animales , Conducta Animal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Femenino , Galactosa , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/genética , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Proteína Oncogénica v-akt/efectos de los fármacos , Proteína Oncogénica v-akt/genética , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Tauopatías/inducido químicamente , Tauopatías/psicología
3.
Cell Mol Neurobiol ; 41(6): 1339-1354, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32696288

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder, in which amyloid precursor protein (APP) misprocessing and tau protein hyperphosphorylation are well-established pathogenic cascades. Despite extensive considerations, the central mediator of neuronal cell death upon AD remains under debate. Therefore, we examined the direct interplay between tauopathy and amyloidopathy processes. We employed primary culture neurons and examined pathogenic P-tau and Aß oligomers upon hypoxia treatment by immunofluorescence and immunoblotting. We observed both tauopathy and amyloidopathy processes upon the hypoxia condition. We also applied Aß1-42 or P-tau onto primary cultured neurons. We overexpressed P-tau in SH-SY5Y cells and found Aß accumulation. Furthermore, adult male rats received Aß1-42 or pathogenic P-tau in the dorsal hippocampus and were examined for 8 weeks. Learning and memory performance, as well as anxiety behaviors, were assessed by Morris water maze and elevated plus-maze tests. Both Aß1-42 and pathogenic P-tau significantly induced learning and memory deficits and enhanced anxiety behavior after treatment 2 weeks. Aß administration induced robust tauopathy distribution in the cortex, striatum, and corpus callosum as well as CA1. On the other hand, P-tau treatment developed Aß oligomers in the cortex and CA1 only. Our findings indicate that Aß1-42 and pathogenic P-tau may induce each other and cause almost identical neurotoxicity in a time-dependent manner, while tauopathy seems to be more distributable than amyloidopathy.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Angiopatía Amiloide Cerebral/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Tauopatías/metabolismo , Proteínas tau/metabolismo , Proteínas tau/toxicidad , Péptidos beta-Amiloides/administración & dosificación , Animales , Línea Celular Tumoral , Células Cultivadas , Angiopatía Amiloide Cerebral/inducido químicamente , Angiopatía Amiloide Cerebral/patología , Femenino , Humanos , Masculino , Ratones , Microinyecciones/métodos , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Wistar , Tauopatías/inducido químicamente , Tauopatías/patología , Proteínas tau/administración & dosificación
4.
Neurotherapeutics ; 17(4): 1878-1896, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32959271

RESUMEN

The molecular mechanism of Alzheimer-like cognitive impairment induced by manganese (Mn) exposure has not yet been fully clarified, and there are currently no effective interventions to treat neurodegenerative lesions related to manganism. Protein phosphatase 2 A (PP2A) is a major tau phosphatase and was recently identified as a potential therapeutic target molecule for neurodegenerative diseases; its activity is directed by the methylation status of the catalytic C subunit. Methionine is an essential amino acid, and its downstream metabolite S-adenosylmethionine (SAM) participates in transmethylation pathways as a methyl donor. In this study, the neurotoxic mechanism of Mn and the protective effect of methionine were evaluated in Mn-exposed cell and rat models. We show that Mn-induced neurotoxicity is characterized by PP2Ac demethylation accompanied by abnormally decreased LCMT-1 and increased PME-1, which are associated with tau hyperphosphorylation and spatial learning and memory deficits, and that the poor availability of SAM in the hippocampus is likely to determine the loss of PP2Ac methylation. Importantly, maintenance of local SAM levels through continuous supplementation with exogenous methionine, or through specific inhibition of PP2Ac demethylation by ABL127 administration in vitro, can effectively prevent tau hyperphosphorylation to reduce cellular oxidative stress, apoptosis, damage to cell viability, and rat memory deficits in cell or animal Mn exposure models. In conclusion, our data suggest that SAM and PP2Ac methylation may be novel targets for the treatment of Mn poisoning and neurotoxic mechanism-related tauopathies.


Asunto(s)
Intoxicación por Manganeso/metabolismo , Manganeso/toxicidad , Metionina/metabolismo , Proteína Fosfatasa 2/metabolismo , Tauopatías/inducido químicamente , Tauopatías/metabolismo , Animales , Línea Celular Tumoral , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Intoxicación por Manganeso/patología , Metilación/efectos de los fármacos , Ratones , Ratas , Ratas Sprague-Dawley , Tauopatías/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-32079163

RESUMEN

Tauopathies are a disease group characterized by either pathological accumulation or release of fragments of hyperphosphorylated tau proteins originating from the central nervous system. The tau hypotheses of Parkinson's and Alzheimer's diseases contain a clinically diverse spectrum of tauopathies. Studies of case records of various tauopathies may reveal clinical phenotype characteristics of the disease. In addition, improved understanding of different tauopathies would disclose environmental factors, such as xenobiotics and trace metals, that can precipitate or modify the progression of the disorder. Important for diagnostics and monitoring of these disorders is a further development of adequate biomarkers, including refined neuroimaging, or proteomics. Our goal is to provide an in-depth review of the current literature regarding the pathophysiological roles of tau proteins and the pathogenic factors leading to various tauopathies, with the perspective of future advances in potential therapeutic strategies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Xenobióticos , Enfermedad de Alzheimer/inducido químicamente , Humanos , Metales , Tauopatías/inducido químicamente , Xenobióticos/toxicidad , Proteínas tau
6.
J Alzheimers Dis ; 70(4): 1025-1040, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31306122

RESUMEN

Recent studies demonstrate that peripheral amylin treatment reduces pathology in mouse models of Alzheimer's disease (AD). However, soluble and aggregated amylin are distinct species; while amylin is a physiological neuropeptide, amylin aggregation is a pathological factor for diabetes. We thus hypothesized that because of their similarity in secondary structures, amylin antagonizes amyloid-ß peptide (Aß)-induced AD pathology in neurons with a dose-dependent pattern. To test the hypothesis, we conducted both in vitro and in vivo experiments with different doses of amylin and with its analog, pramlintide. Here we report that a high concentration of either Aß or amylin alone induced tau phosphorylation (pTau) in primary neurons. Interestingly, with a low concentration, amylin had direct effects to reverse the Aß-induced pTau, as well as damaged neuronal synapses and neurite disorganization. However, when the concentration was high (10.24 µM), amylin lost the effects against the Aß-induced cellular AD pathology and, together with Aß, worsened tauopathy in neurons. In the 5XFAD AD mouse model, daily peripheral amylin treatment with a low dose (200 µg/kg) more effectively reduced amyloid burden, and increased synapse, but with a high dose (800 µg/kg), it more effectively reduced tauopathy. Correspondingly, amylin treatment improved learning and memory in these mice. It demonstrates that amylin has a dose-dependent U-shape effect against AD pathogenesis. Within a physiological range, amylin is a neuroprotective hormone against AD in neurons; but when both Aß and amylin concentrations are elevated, imbalance of Aß and amylin may contribute to brain AD pathogenesis.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Sinapsis/efectos de los fármacos , Tauopatías/inducido químicamente , Tauopatías/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/uso terapéutico , Ratones , Ratones Transgénicos , Neuronas/patología , Embarazo , Ratas , Ratas Wistar , Sinapsis/patología , Tauopatías/patología
7.
Neuroimage ; 189: 180-191, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30630081

RESUMEN

White matter abnormalities, revealed by Diffusion Tensor Imaging (DTI), are observed in patients with Alzheimer's Disease (AD), representing neural network deficits that underlie gradual cognitive decline in patients. However, how DTI changes related to the development of Amyloid beta (Aß) and tau pathology, two key hallmarks of AD, remain elusive. We hypothesized that tauopathy induced by Aß could initiate an axonal degeneration, leading to DTI-detectable white matter abnormalities. We utilized the visual system of the transgenic p301L tau mice as a model system. Aß was injected in Lateral Geniculate Nucleus (LGN), where the Retinal Ganglion Cell (RGC) axons terminate. Longitudinal DTI was conducted to detect changes in the optic tract (OT) and optic nerve (ON), containing the distal and proximal segments of RGC axons, respectively. Our results showed DTI changes in OT (significant 13.2% reduction in axial diffusion, AxD vs. vehicle controls) followed by significant alterations in ON AxD and fractional anisotropy, FA. Histology data revealed loss of synapses, RGC axons and cell bodies resulting from the Aß injection. We further tested whether microtubule-stabilizing compound Epothilone D (EpoD) could ameliorate the damage. EpoD co-treatment with Aß was sufficient to prevent Aß-induced axon and cell loss. Using an acute injection paradigm, our data suggest that EpoD may mediate its protective effect by blocking localized, acute Aß-induced tau phosphorylation. This study demonstrates white matter disruption resulting from localized Aß, the importance of tau pathology induction to changes in white matter connectivity, and the use of EpoD as a potential therapeutic avenue to prevent the axon loss in AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Epotilonas/farmacología , Cuerpos Geniculados/efectos de los fármacos , Degeneración Nerviosa , Fragmentos de Péptidos/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Tauopatías , Moduladores de Tubulina/farmacología , Sustancia Blanca , Péptidos beta-Amiloides/administración & dosificación , Animales , Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Epotilonas/administración & dosificación , Ratones , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Fragmentos de Péptidos/administración & dosificación , Tauopatías/inducido químicamente , Tauopatías/diagnóstico por imagen , Tauopatías/tratamiento farmacológico , Tauopatías/patología , Moduladores de Tubulina/administración & dosificación , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/efectos de los fármacos
8.
Neurotox Res ; 35(4): 987-992, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30362086

RESUMEN

Tauopathy is a pathological hallmark of many neurodegenerative diseases. It is characterized by abnormal aggregates of pathological phosphotau and somatodendritic redistribution. One suggested strategy for treating tauopathy is to stimulate autophagy, hence, getting rid of these pathological protein aggregates. One key controller of autophagy is mTOR. Since stimulation of mTOR leads to inhibition of autophagy, inhibitors of mTOR will cause stimulation of autophagy process. In this report, tauopathy was induced in mice using annonacin. Blocking of mTOR was achieved through stereotaxic injection of siRNA against mTOR. The behavioral and immunohistochemical evaluation revealed the development of tauopathy model as proven by deterioration of behavioral performance in open field test and significant tau aggregates in annonacin-treated mice. Blocking of mTOR revealed significant clearance of tau aggregates in the injected side; however, tau expression was not affected by mTOR blockage.


Asunto(s)
Encéfalo/patología , Neuronas/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tauopatías/patología , Animales , Autofagia , Encéfalo/metabolismo , Furanos , Lactonas , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Agregación Patológica de Proteínas/metabolismo , ARN Interferente Pequeño/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Tauopatías/inducido químicamente , Tauopatías/metabolismo
9.
Ecotoxicol Environ Saf ; 169: 301-305, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30458396

RESUMEN

Epidemiological and toxicological studies have shown that ambient fine particulate matter (PM2.5) is a healthy risk factor for neurodegenerative diseases. Hyperphosphorylated tau is the common feature of numerous neurodegenerative diseases known as tauopathy, which could be inhibited by insulin stimulation. However, the effects of PM2.5 on tau protein injury by disturbing the insulin signaling pathway still need to be illuminated. In present study, male C57BL/6 J mice were administered with PM2.5 to determine whether PM2.5 inhalation can induce tauopathy via the insulin resistance (IR) related pathway (IRS-1/AKT/GSK-3ß signaling pathway). The results showed that PM2.5 treatment induced the generation of phosphorylated tau (P-tau) and contributed to the development of tauopathy because of the insulin signaling disorders in insulin targeting organs. As expected, the occurrence of central and peripheral IR and accompanying hyperinsulinemia aggravated the disturbance of the IRS-1/AKT/GSK-3ß signaling pathway. These observations indicated that PM2.5 exposure led to neurodegenerative tau lesion, and insulin signaling pathway might be a potential therapeutic target for tauopathy.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Insulina/metabolismo , Material Particulado/toxicidad , Tauopatías/inducido químicamente , Proteínas tau/metabolismo , Animales , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Fosforilación , Transducción de Señal/efectos de los fármacos , Tauopatías/metabolismo
10.
Acta Neuropathol Commun ; 6(1): 103, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30270813

RESUMEN

Chemotherapy-induced cognitive impairment (CICI) is a commonly reported neurotoxic side effect of chemotherapy, occurring in up to 75% cancer patients. CICI manifests as decrements in working memory, executive functioning, attention, and processing speed, and greatly interferes with patients' daily performance and quality of life. Currently no treatment for CICI has been approved by the US Food and Drug Administration. We show here that treatment with a brain-penetrating histone deacetylase 6 (HDAC6) inhibitor for two weeks was sufficient to fully reverse cisplatin-induced cognitive impairments in male mice, as demonstrated in the Y-maze test of spontaneous alternation, the novel object/place recognition test, and the puzzle box test. Normalization of cognitive impairment was associated with reversal of cisplatin-induced synaptosomal mitochondrial deficits and restoration of synaptic integrity. Mechanistically, cisplatin induced deacetylation of the microtubule protein α-tubulin and hyperphosphorylation of the microtubule-associated protein tau. These cisplatin-induced changes were reversed by HDAC6 inhibition. Our data suggest that inhibition of HDAC6 restores microtubule stability and reverses tau phosphorylation, leading to normalization of synaptosomal mitochondrial function and synaptic integrity and thereby to reversal of CICI. Remarkably, our results indicate that short-term daily treatment with the HDAC6 inhibitor was sufficient to achieve prolonged reversal of established behavioral, structural and functional deficits induced by cisplatin. Because the beneficial effects of HDAC6 inhibitors as add-ons to cancer treatment have been demonstrated in clinical trials, selective targeting of HDAC6 with brain-penetrating inhibitors appears a promising therapeutic approach for reversing chemotherapy-induced neurotoxicity while enhancing tumor control.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Disfunción Cognitiva , Inhibidores Enzimáticos/uso terapéutico , Histona Desacetilasa 6/metabolismo , Tauopatías/enzimología , Animales , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/enzimología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Proteínas Fluorescentes Verdes/metabolismo , Histona Desacetilasa 6/ultraestructura , Ácidos Hidroxámicos/sangre , Ácidos Hidroxámicos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Pirimidinas/sangre , Pirimidinas/uso terapéutico , Proteínas Recombinantes de Fusión/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Sinaptosomas/patología , Sinaptosomas/ultraestructura , Tauopatías/inducido químicamente , Tauopatías/tratamiento farmacológico , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Proteínas tau/metabolismo
11.
J Alzheimers Dis ; 64(2): 617-630, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29914030

RESUMEN

Hyperphosphorylated tau protein is a key pathology in Alzheimer's disease (AD), frontotemporal dementia, chronic traumatic encephalopathy, and Parkinson's disease. The essential trace element zinc exacerbates tauopathy in vitro as well as in a Drosophila model of AD. However, the interaction has never been assessed behaviorally or biochemically in mammals. Zinc supplementation is prevalent in society, finding use as a treatment for macular degeneration and cataracts, and is also taken as an immune system booster with high levels appearing in multivitamins marketed toward the elderly. Using a transgenic mouse model that contains the human gene for tau protein (P301L), we assessed the effects of excess chronic zinc supplementation on tau pathology. Behavioral tests included nest building, circadian rhythm, Morris Water Maze, fear conditioning, and open field. Biochemically, total tau and Ser396 phosphorylation were assessed using western blot. Number of tangles were assessed by Thioflavin-S and free zinc levels were assessed by Zinpyr-1. Tau mice demonstrated behavioral deficits compared to control mice. Zinc supplementation exacerbated tauopathic deficits in circadian rhythm, nesting behavior, and Morris Water Maze. Biochemically, zinc-supplemented tau mice showed increased phosphorylation at pSer396. Zinc supplementation in tau mice also increased tangle numbers in the hippocampus while decreasing free-zinc levels, demonstrating that tangles were sequestering zinc. These results show that zinc intensified the deficits in behavior and biochemistry caused by tau.


Asunto(s)
Tauopatías/inducido químicamente , Tauopatías/genética , Zinc/toxicidad , Proteínas tau/metabolismo , Animales , Ritmo Circadiano/efectos de los fármacos , Ritmo Circadiano/genética , Condicionamiento Psicológico/efectos de los fármacos , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Femenino , Fluoresceínas/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Comportamiento de Nidificación/efectos de los fármacos , Tauopatías/fisiopatología , Proteínas tau/genética
12.
J Alzheimers Dis ; 63(1): 13-33, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29630551

RESUMEN

With predictions showing that 131.5 million people worldwide will be living with dementia by 2050, an understanding of the molecular mechanisms underpinning disease is crucial in the hunt for novel therapeutics and for biomarkers to detect disease early and/or monitor disease progression. The metabolism of the microtubule-associated protein tau is altered in different dementias, the so-called tauopathies. Tau detaches from microtubules, aggregates into oligomers and neurofibrillary tangles, which can be secreted from neurons, and spreads through the brain during disease progression. Post-translational modifications exacerbate the production of both oligomeric and soluble forms of tau, with proteolysis by a range of different proteases being a crucial driver. However, the impact of tau proteolysis on disease progression has been overlooked until recently. Studies have highlighted that proteolytic fragments of tau can drive neurodegeneration in a fragment-dependent manner as a result of aggregation and/or transcellular propagation. Proteolytic fragments of tau have been found in the cerebrospinal fluid and plasma of patients with different tauopathies, providing an opportunity to develop these fragments as novel disease progression biomarkers. A range of therapeutic strategies have been proposed to halt the toxicity associated with proteolysis, including reducing protease expression and/or activity, selectively inhibiting protease-substrate interactions, and blocking the action of the resulting fragments. This review highlights the importance of tau proteolysis in the pathogenesis of tauopathies, identifies putative sites during tau fragment-mediated neurodegeneration that could be targeted therapeutically, and discusses the potential use of proteolytic fragments of tau as biomarkers for different tauopathies.


Asunto(s)
Biomarcadores/metabolismo , Fragmentos de Péptidos/toxicidad , Proteolisis , Tauopatías , Proteínas tau/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Tauopatías/inducido químicamente , Tauopatías/metabolismo , Tauopatías/patología
13.
ACS Chem Neurosci ; 8(12): 2734-2745, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28837764

RESUMEN

In the present study, a cell penetrating peptide (CPP)-amyloid conjugate was prepared (T-peptide), where the amyloid-forming sequence was homologous to a nucleating sequence from human Tau protein (306VQIVYK311). Kinetic and biophysical studies showed the peptide formed long-lived oligomers which were taken up by endocytosis and localized in perinuclear vesicles and in the cytoplasm of murine hippocampal neuroblastoma cells and human HeLa cells. Thioflavin S (ThS) staining of amyloid colocalized with pathological phosphorylated Tau, suggesting that the peptide was able to seed endogenous wild-type Tau. Subsequent experiments showed that aggregates present in the lysosomes mediated lysosome membrane permeability (LMP). We observed a decrease in total Tau, irrespective of phosphorylation state, consistent with Tau fragmentation by lysosomal proteases. We found cytotoxicity of T-peptide could be abrogated by inhibitors of lysosomal hydrolases and caspases, consistent with a model where Tau fragments processed by the lysosome leak into the cytoplasm and induce toxicity in subsequent downstream steps. It is our hope that the T-peptide system may prove amenable to the evaluation of small molecule inhibitors of cytotoxicity, especially those which target either Tau aggregation or the lysosomal/autophagy system.


Asunto(s)
Péptidos de Penetración Celular/farmacocinética , Modelos Animales de Enfermedad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Tauopatías/inducido químicamente , Tauopatías/metabolismo , Proteínas tau/metabolismo , Amiloide , Animales , Línea Celular , Membrana Celular , Péptidos de Penetración Celular/administración & dosificación , Células HeLa , Humanos , Ratones , Neuronas/patología , Tauopatías/patología
14.
Biochem Biophys Res Commun ; 483(1): 585-589, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-27998769

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia, characterized by progressive loss of memory and other cognitive functions. The cognitive impairment in patients with AD is closely associated with loss of synapses and the formation of neurofibrillary tangles (NFT) containing hyperphosphorylated tau in the hippocampus. Effective treatment for AD is still not available. In this study, the sequence comprising of residues 50-71 in the N-terminal region of tau, containing theoretically predicted B- and T-cell epitopes in close proximity to pathologically relevant phospho-serine (residue 68) and phospho-threonine (residues 69, 71) was selected as a potential immunotherapeutic peptide. This 22-residue long phospho-peptide (50TPTEDGSEEPGSETSDAKpSpTPpT71) was custom synthesized and its therapeutic potential was tested in experimental rats. For this purpose, adult Sprague-Dawley rats were intranasally treated with okadaic acid (OA), a selective inhibitor of protein phosphatase PP2A. Within a day of OA administration, these rats showed marked impairment in cognitive functions with a significant increase in p-tau/t-tau ratio in the hippocampal homogenates. Passive immunization studies conducted in these OA treated rats with polyclonal anti-phospho-peptide antibodies resulted in a significant improvement in learning and memory functions in Barne's maze task. Further, p-tau levels in the hippocampal homogenates were reduced. In addition, these antibodies effectively prevented the aggregation of recombinant tau in vitro. These results demonstrate that targeting N-terminal region of tau harbouring the phospho-residue cluster 68-71 would be beneficial and may present an effective therapeutic opportunity for AD and other tauopathies.


Asunto(s)
Inmunización/métodos , Memoria Espacial , Tauopatías/terapia , Proteínas tau/química , Animales , Modelos Animales de Enfermedad , Epítopos/química , Femenino , Hipocampo/metabolismo , Masculino , Ácido Ocadaico/química , Péptidos/química , Fosforilación , Dominios Proteicos , Proteína Fosfatasa 2/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Tauopatías/inducido químicamente
15.
Chemosphere ; 165: 248-256, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27657817

RESUMEN

Air pollution has been evidenced as a risk factor for neurodegenerative tauopathies. NO2, a primary component of air pollution, is negatively linked to neurodegenerative disorders, but its independent and direct association with tau lesion remains to be elucidated. Considering the fact that the insulin signaling pathway can be targeted by air pollutants and regulate tau function, this study focused on the role of insulin signaling in this NO2-induced tauopathy. Using a dynamic inhalation treatment, we demonstrated that exposure to NO2 induced a disruption of insulin signaling in skeletal muscle, liver, and brain, with associated p38 MAPK and/or JNK activation. We also found that in parallel with these kinase signaling cascades, the compensatory hyperinsulinemia triggered by whole-body insulin resistance (IR) further attenuated the IRS-1/AKT/GSK-3ß signaling pathway in the central nervous system, which consequently increased the phosphorylation of tau and reduced the expression of synaptic proteins that contributed to the development of the tau pathology. These findings provide new insight into the possible mechanisms involved in the etiopathogenesis of NO2-induced tauopathy, suggesting that the targeting of insulin signaling may be a promising therapeutic strategy to prevent this disease.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Sistema Nervioso Central/patología , Exposición a Riesgos Ambientales , Inhalación , Resistencia a la Insulina , Insulina/metabolismo , Dióxido de Nitrógeno/toxicidad , Tauopatías/inducido químicamente , Contaminación del Aire , Animales , Encéfalo/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hiperinsulinismo/inducido químicamente , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas tau/metabolismo
16.
Neurobiol Dis ; 73: 83-95, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25220759

RESUMEN

Neurofibrillary tangles composed of hyperphosphorylated fibrillized tau are found in numerous tauopathies including Alzheimer's disease. Increasing evidence suggests that tau pathology can be transmitted from cell-to-cell; however the mechanisms involved in the initiation of tau fibrillization and spreading of disease linked to progression of tau pathology are poorly understood. We show here that intracerebral injections of preformed synthetic tau fibrils into the hippocampus or frontal cortex of young tau transgenic mice expressing mutant human P301L tau induces tau hyperphosphorylation and aggregation around the site of injection, as well as a time-dependent propagation of tau pathology to interconnected brain areas distant from the injection site. Furthermore, we show that the tau pathology as a consequence of injection of tau preformed fibrils into the hippocampus induces selective loss of CA1 neurons. Together, our data confirm previous studies on the seeded induction and the spreading of tau pathology in a different tau transgenic mouse model and reveals neuronal loss associated with seeded tau pathology in tau transgenic mouse brain. These results further validate the utility of the tau seeding model in studying disease transmission, and provide a more complete in vivo tauopathy model with associated neurodegeneration which can be used to investigate the mechanisms involved in tau aggregation and spreading, as well as aid in the search for disease modifying treatments for Alzheimer's disease and related tauopathies.


Asunto(s)
Tauopatías , Proteínas tau/administración & dosificación , Proteínas tau/genética , Factores de Edad , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Ovillos Neurofibrilares/metabolismo , Tauopatías/inducido químicamente , Tauopatías/genética , Tauopatías/patología , Proteínas tau/química
17.
Genet Mol Res ; 13(3): 6213-27, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25158248

RESUMEN

The amyloid C-terminal fragment (ßCTF) of the amyloid precursor protein (APP) is the cleaved component of APP by beta secretase-1 (BACE1), which shows similar neurotoxicity as amyloid beta (Aß) in many ways. Evidence suggested that in addition to Aß, ßCTF might also participate in the pathogenesis of Alzheimer's disease (AD). In recent years, the relationship between ßCTF processing and hyperphosphorylated tau has attracted increasing research attention. In this study, we established an animal model of tau hyperphosphorylation with okadaic acid (OA) treatment, and analyzed ßCTF processing in vivo. The ßCTF level was found to increase in neurons, which was most likely caused by the induction of OA and BACE1 overexpression. Furthermore, these results provide the first evidence that ßCTF can predominately accumulate in the axons of neurons in a hyperphosphorylated tau state in vivo, and suggested that the redistribution of ßCTF is involved in the pathogenesis of AD. These results indicate that BACE1 could be a therapeutic target of AD by affecting the processing of ßCTF.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Neuronas/metabolismo , Inhibidores de Proteasas/farmacología , Tauopatías/genética , Proteínas tau/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Ácido Ocadaico , Fosforilación , Ratas , Ratas Sprague-Dawley , Técnicas Estereotáxicas , Tauopatías/inducido químicamente , Tauopatías/tratamiento farmacológico , Tauopatías/patología , Proteínas tau/genética
19.
J Alzheimers Dis ; 33(2): 339-48, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22936006

RESUMEN

Misfolded, N- and C-terminally truncated tau protein is the primary constituent of neurofibrillary tangles in brains of patients afflicted with Alzheimer's disease (AD). Intracellular accumulation of misfolded and truncated tau leads to generation of cytotoxic intermediates; transgenic expression of truncated tau leads to neurological deficits, neurofibrillary degeneration, and premature death of animals. Since no cure for AD or other tauopathies is available yet, we tested the possibility for prevention of pathogenic events elicited by tau, via inhibition of its intracellular accumulation. Using a cell model conditionally expressing truncated and misfolding-prone tau protein, we showed that pathogenic forms of tau are degraded via the proteasome. We have also observed that chymotrypsin-like activity of the proteasome was significantly suppressed (a decrease of ∼29.12% in comparison to control cells; p < 0.001) as a consequence of truncated tau expression. Interestingly, the activity of the proteasome was enhanced by geldanamycin, a natural inhibitor of Hsp90. This activation resulted in accelerated degradation of misfolded tau. We suggest that non-toxic inhibitors of Hsp90, especially those which can activate the proteasome, are good candidates for the development of molecules that efficiently counteract the damaging effects of pathologically misfolded proteins.


Asunto(s)
Benzoquinonas/toxicidad , Lactamas Macrocíclicas/toxicidad , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Deficiencias en la Proteostasis/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Quimotripsina/metabolismo , Inhibidores de Cisteína Proteinasa/toxicidad , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Neuroblastoma , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pliegue de Proteína , Deficiencias en la Proteostasis/inducido químicamente , Deficiencias en la Proteostasis/patología , Tauopatías/inducido químicamente , Tauopatías/patología , Transgenes/genética , Proteínas tau/química , Proteínas tau/genética
20.
PLoS One ; 7(1): e30745, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22292029

RESUMEN

SNCA and MAPT genes and environmental factors are important risk factors of Parkinson's disease [PD], the second-most common neurodegenerative disease. The agrichemicals maneb and paraquat selectively target dopaminergic neurons, leading to parkinsonism, through ill-defined mechanisms. In the current studies we have analyzed the ability of maneb and paraquat, separately and together, to induce synucleinopathy and tauopathy in wild type mice. Maneb was ineffective in increasing α-synuclein [α-Syn] or p-Tau levels. By contrast, paraquat treatment of mice resulted in robust accumulation of α-Syn and hyperphosphorylation of Tau in striata, through activation of p-GSK-3ß, a major Tau kinase. Co-treatment with maneb did not enhance the effects of paraquat. Increased hyperacetylation of α-tubulin was observed in paraquat-treated mice, suggesting cytoskeleton remodeling. Paraquat, but not maneb, inhibited soluble proteasomal activity on a peptide substrate but this was not associated with a decreased expression of 26S proteasome subunits. Both paraquat and maneb treatments increased levels of the autophagy inhibitor, mammalian target of rapamycin, mTOR, suggesting impaired axonal autophagy, despite increases in certain autophagic proteins, such as beclin 1 and Agt12. Autophagic flux was also impaired, as ratios of LC3 II to LC3 I were reduced in treated animals. Increased mTOR was also observed in postmortem human PD striata, where there was a reduction in the LC3 II to LC3 I ratio. Heat shock proteins were either increased or unchanged upon paraquat-treatment suggesting that chaperone-mediated autophagy is not hampered by the agrichemicals. These studies provide novel insight into the mechanisms of action of these agrichemicals, which indicate that paraquat is much more toxic than maneb, via its inhibitory effects on proteasomes and autophagy, which lead to accumulation of α-Syn and p-Tau.


Asunto(s)
Autofagia/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Maneb/toxicidad , Paraquat/toxicidad , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Tauopatías/inducido químicamente , Animales , Autofagia/genética , Estudios de Casos y Controles , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Regulación hacia Abajo/efectos de los fármacos , Fungicidas Industriales/toxicidad , Herbicidas/toxicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/efectos de los fármacos , Tauopatías/genética , Tauopatías/patología , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA