Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
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
2.
Neural Plast ; 2019: 6981268, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31285742

RESUMEN

Alzheimer's disease (AD) is a progressive, neurodegenerative disease characterized by loss of synapses and disrupted functional connectivity (FC) across different brain regions. Early in AD progression, tau pathology is found in the locus coeruleus (LC) prior to amyloid-induced exacerbation of clinical symptoms. Here, a tau-seeding model in which preformed synthetic tau fibrils (K18) were unilaterally injected into the LC of P301L mice, equipped with multichannel electrodes for recording EEG in frontal cortical and CA1-CA3 hippocampal areas, was used to longitudinally quantify over 20 weeks of functional network dynamics in (1) power spectra; (2) FC using intra- and intersite phase-amplitude theta-gamma coupling (PAC); (3) coherence, partial coherence, and global coherent network efficiency (Eglob) estimates; and (4) the directionality of functional connectivity using extended partial direct coherence (PDC). A sustained leftward shift in the theta peak frequency was found early in the power spectra of hippocampal CA1 networks ipsilateral to the injection site. Strikingly, hippocampal CA1 coherence and Eglob measures were impaired in K18-treated animals. Estimation of instantaneous EEG amplitudes revealed deficiency in the propagation directionality of gamma oscillations in the CA1 circuit. Impaired PAC strength evidenced by decreased modulation of the theta frequency phase on gamma frequency amplitude further confirms impairments of the neural CA1 network. The present results demonstrate early dysfunctional hippocampal networks, despite no spreading tau pathology to the hippocampus and frontal cortex. The ability of the K18 seed in the brainstem LC to elicit such robust functional alterations in distant hippocampal structures in the absence of pathology challenges the classic view that tau pathology spread to an area is necessary to elicit functional impairments in that area.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Locus Coeruleus/fisiopatología , Red Nerviosa/fisiopatología , Proteínas tau/toxicidad , Enfermedad de Alzheimer/genética , Animales , Electroencefalografía/métodos , Fenómenos Electrofisiológicos/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas Estereotáxicas , Factores de Tiempo , Proteínas tau/administración & dosificación
3.
J Neurosci ; 37(47): 11485-11494, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-28986461

RESUMEN

Neurodegenerative proteinopathies characterized by intracellular aggregates of tau proteins, termed tauopathies, include Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD) with tau pathology (FTLD-tau), and related disorders. Pathological tau proteins derived from human AD brains (AD-tau) act as proteopathic seeds that initiate the templated aggregation of soluble tau upon intracerebral injection into tau transgenic (Tg) and wild-type mice, thereby modeling human tau pathology. In this study, we found that aged Tg mice of both sexes expressing human tau proteins harboring a pathogenic P301L MAPT mutation labeled with green fluorescent protein (T40PL-GFP Tg mouse line) exhibited hyperphosphorylated tau mislocalized to the somatodentritic domain of neurons, but these mice did not develop de novo insoluble tau aggregates, which are characteristic of human AD and related tauopathies. However, intracerebral injections of either T40PL preformed fibrils (PFFs) or AD-tau seeds into T40PL-GFP mice induced abundant intraneuronal pathological inclusions of hyperphosphorylated T40PL-GFP. These injections of pathological tau resulted in the propagation of tau pathology from the injection site to neuroanatomically connected brain regions, and these tau inclusions consisted of both T40PL-GFP and WT endogenous mouse tau. Primary neurons cultured from the brains of neonatal T40PL-GFP mice provided an informative in vitro model for examining the uptake and localization of tau PFFs. These findings demonstrate the seeded aggregation of T40PL-GFP in vivo by synthetic PFFs and human AD-tau and the utility of this system to study the neuropathological spread of tau aggregates.SIGNIFICANCE STATEMENT The stereotypical spread of pathological tau protein aggregates have recently been attributed to the transmission of proteopathic seeds. Despite the extensive use of transgenic mouse models to investigate the propagation of tau pathology in vivo, details of the aggregation process such as the early seeding events leading to new tau pathology have remained elusive. This study validates the use of GFP-labeled tau expressed by neurons in vivo and in vitro as models for investigating mechanisms underlying the seeded transmission of tau pathology as well as tau-focused drug discovery to identify disease-modifying therapies for AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/toxicidad , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , Mutación , Neuronas/metabolismo , Neuronas/patología , Proteínas Recombinantes , Proteínas tau/administración & dosificación , Proteínas tau/genética , Proteínas tau/metabolismo
4.
Cell Mol Life Sci ; 72(23): 4613-32, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26070304

RESUMEN

Patients suffering from tauopathies including frontotemporal dementia (FTD) and Alzheimer's disease (AD) present with intra-neuronal aggregation of microtubule-associated protein Tau. During the disease process, Tau undergoes excessive phosphorylation, dissociates from microtubules and aggregates into insoluble neurofibrillary tangles (NFTs), accumulating in the soma. While many aspects of the disease pathology have been replicated in transgenic mouse models, a region-specific non-transgenic expression model is missing. Complementing existing models, we here report a novel region-specific approach to modelling Tau pathology. Local co-administration of the pore-former polymeric 1,3-alkylpyridinium salts (Poly-APS) extracted from marine sponges, and synthetic full-length 4R recombinant human Tau (hTau) was performed in vitro and in vivo. At low doses, Poly-APS was non-toxic and cultured cells exposed to Poly-APS (0.5 µg/ml) and hTau (1 µg/ml; ~22 µM) had normal input resistance, resting-state membrane potentials and Ca(2+) transients induced either by glutamate or KCl, as did cells exposed to a low concentration of the phosphatase inhibitor Okadaic acid (OA; 1 nM, 24 h). Combined hTau loading and phosphatase inhibition resulted in a collapse of the membrane potential, suppressed excitation and diminished glutamate and KCl-stimulated Ca(2+) transients. Stereotaxic infusions of Poly-APS (0.005 µg/ml) and hTau (1 µg/ml) bilaterally into the dorsal hippocampus at multiple sites resulted in hTau loading of neurons in rats. A separate cohort received an additional 7-day minipump infusion of OA (1.2 nM) intrahippocampally. When tested 2 weeks after surgery, rats treated with Poly-APS+hTau+OA presented with subtle learning deficits, but were also impaired in cognitive flexibility and recall. Hippocampal plasticity recorded from slices ex vivo was diminished in Poly-APS+hTau+OA subjects, but not in other treatment groups. Histological sections confirmed the intracellular accumulation of hTau in CA1 pyramidal cells and along their processes; phosphorylated Tau was present only within somata. This study demonstrates that cognitive, physiological and pathological symptoms reminiscent of tauopathies can be induced following non-mutant hTau delivery into CA1 in rats, but functional consequences hinge on increased Tau phosphorylation. Collectively, these data validate a novel model of locally infused recombinant hTau protein as an inducer of Tau pathology in the hippocampus of normal rats; future studies will provide insights into the pathological spread and maturation of Tau pathology.


Asunto(s)
Hipocampo/citología , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Polímeros/administración & dosificación , Compuestos de Piridinio/administración & dosificación , Proteínas tau/farmacología , Animales , Conducta Animal/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Electrofisiología/métodos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Discapacidades para el Aprendizaje/tratamiento farmacológico , Masculino , Neuronas/metabolismo , Neuronas/fisiología , Fosforilación , Polímeros/farmacología , Compuestos de Piridinio/farmacología , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Tauopatías/metabolismo , Proteínas tau/administración & dosificación , Proteínas tau/metabolismo
5.
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
6.
J Neurosci ; 31(37): 13110-7, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21917794

RESUMEN

Although tau is a cytoplasmic protein, it is also found in brain extracellular fluids, e.g., CSF. Recent findings suggest that aggregated tau can be transferred between cells and extracellular tau aggregates might mediate spread of tau pathology. Despite these data, details of whether tau is normally released into the brain interstitial fluid (ISF), its concentration in ISF in relation to CSF, and whether ISF tau is influenced by its aggregation are unknown. To address these issues, we developed a microdialysis technique to analyze monomeric ISF tau levels within the hippocampus of awake, freely moving mice. We detected tau in ISF of wild-type mice, suggesting that tau is released in the absence of neurodegeneration. ISF tau was significantly higher than CSF tau and their concentrations were not significantly correlated. Using P301S human tau transgenic mice (P301S tg mice), we found that ISF tau is fivefold higher than endogenous murine tau, consistent with its elevated levels of expression. However, following the onset of tau aggregation, monomeric ISF tau decreased markedly. Biochemical analysis demonstrated that soluble tau in brain homogenates decreased along with the deposition of insoluble tau. Tau fibrils injected into the hippocampus decreased ISF tau, suggesting that extracellular tau is in equilibrium with extracellular or intracellular tau aggregates. This technique should facilitate further studies of tau secretion, spread of tau pathology, the effects of different disease states on ISF tau, and the efficacy of experimental treatments.


Asunto(s)
Envejecimiento/metabolismo , Líquido Extracelular/metabolismo , Hipocampo/metabolismo , Microdiálisis/métodos , Proteínas tau/genética , Proteínas tau/metabolismo , Envejecimiento/líquido cefalorraquídeo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Modelos Neurológicos , Solubilidad , Proteínas tau/administración & dosificación , Proteínas tau/líquido cefalorraquídeo , Proteínas tau/química
7.
Acta Neuropathol Commun ; 9(1): 150, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34503586

RESUMEN

Dystrophic neuronal processes harboring neuritic plaque (NP) tau pathology are found in association with Aß plaques in Alzheimer's disease (AD) brain. Microglia are also in proximity to these plaques and microglial gene variants are known risk factors in AD, including loss-of-function variants of TREM2. We have further investigated the role of Aß plaque-associated microglia in 5XFAD mice in which NP tau pathology forms after intracerebral injection of AD brain-derived pathologic tau (AD-tau), focusing on the consequences of reduced TREM2 expression and microglial depletion after treatment with the colony-stimulating factor 1 (CSFR1) inhibitor, PLX3397. Young 5XFAD mice treated with PLX3397 had a large reduction of brain microglia, including cortical plaque-associated microglia, with a significant reduction of Aß plaque burden in the cortex. A corresponding decrease in cortical APP-positive dystrophic processes and NP tau pathology were observed after intracerebral AD-tau injection in the PLX3397-treated 5XFAD mice. Consistent with prior reports, 5XFAD × TREM2-/- mice showed a significant reduction of plaque-associated microglial, whereas 5XFAD × TREM2+/- mice had significantly more plaque-associated microglia than 5XFAD × TREM2-/- mice. Nonetheless, AD-tau injected 5XFAD × TREM2+/- mice showed greatly increased AT8-positive NP tau relative to 5XFAD × TREM2+/+ mice. Expression profiling revealed that 5XFAD × TREM2+/- mice had a disease-associated microglial (DAM) gene expression profile in the brain that was generally intermediate between 5XFAD × TREM2+/+ and 5XFAD × TREM2-/- mice. Microarray analysis revealed significant differences in cortical and hippocampal gene expression between AD-tau injected 5XFAD × TREM2+/- and 5XFAD × TREM2-/- mice, including pathways linked to microglial function. These data suggest there is not a simple correlation between the extent of microglia plaque interaction and plaque-associated neuritic damage. Moreover, the differences in gene expression and microglial phenotype between TREM2+/- and TREM2-/- mice suggest that the former may better model the single copy TREM2 variants associated with AD risk.


Asunto(s)
Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Placa Amiloide/metabolismo , Receptores Inmunológicos/deficiencia , Proteínas tau/toxicidad , Animales , Femenino , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Placa Amiloide/inducido químicamente , Placa Amiloide/genética , Receptores Inmunológicos/genética , Proteínas tau/administración & dosificación
8.
J Alzheimers Dis ; 77(2): 551-568, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32675411

RESUMEN

BACKGROUND: Neuronal accumulation of hyperphosphorylated and truncated tau aggregates is one of the major defining factors and key drivers of neurodegeneration in Alzheimer's disease and other tauopathies. OBJECTIVE: We developed an AAV-induced model of tauopathy mediated by human truncated tau protein without familial frontotemporal dementia-related mutations to study tau propagation and the functional consequences of tau pathology. METHODS: We performed targeted transductions of the hippocampus or entorhinal cortex in adult mice followed by histological analysis to study the progression of hippocampal tau pathology and tau spreading. We performed behavioral analysis of mice with AAV-induced hippocampal tau pathology. RESULTS: AAV-induced hippocampal tau pathology was characterized by tau hyperphosphorylation (AT8 positivity), sarkosyl insolubility, and the presence of neurofibrillary tangles. AAV-induced tau pathology was associated with microgliosis and hypertrophic astrocytes in the absence of cognitive deficits. Additionally, the co-expression of mCherry fluorescent protein and human truncated tau enabled us to detect both local spreading of human tau and spreading from the entorhinal cortex to the synaptically connected dentate gyrus. CONCLUSION: Targeted delivery of AAV with truncated tau protein into subcortical and cortical structures of mammalian brains represents an efficient approach for creating temporally and spatially well-defined tau pathology suitable for in vivo studies of tau propagation and neuronal circuit deficits in Alzheimer's disease.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Mutación , Neuronas/virología , Tauopatías/virología , Proteínas tau/administración & dosificación , Adenoviridae/genética , Animales , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
9.
Neurosci Biobehav Rev ; 112: 1-27, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31996301

RESUMEN

Alzheimer's disease (AD) is characterized neuropathologically by progressive neurodegeneration and by the presence of amyloid plaques and neurofibrillary tangles. These plaques and tangles are composed, respectively, of amyloid-beta (Aß) and tau proteins. While long recognized as hallmarks of AD, it remains unclear what causes the formation of these insoluble deposits. One theory holds that prion-like templated misfolding of Aß and tau induces these proteins to form pathological aggregates, and propagation of this misfolding causes the stereotyped progression of pathology commonly seen in AD. Supporting this theory, numerous studies have been conducted in which aggregated Aß, tau, or α-synuclein is injected intracerebrally into pathology-free host animals, resulting in robust formation of pathology. Here, we review this literature, focusing on in vivo intracerebral seeding of Aß and tau in mice. We compare the results of these experiments to what is known about the seeding and spread of α-synuclein pathology, and we discuss how this research informs our understanding of the factors underlying the onset, progression, and outcomes of proteinaceous pathologies.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades por Prión/inducido químicamente , alfa-Sinucleína/farmacología , Proteínas tau/farmacología , Péptidos beta-Amiloides/administración & dosificación , Animales , Ratones , alfa-Sinucleína/administración & dosificación , Proteínas tau/administración & dosificación
10.
Cell Res ; 30(1): 70-87, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31649329

RESUMEN

Lewy pathology, composed of α-Synuclein (α-Syn) inclusions, a hallmark of Parkinson's disease (PD), progressively spreads from the enteric nervous system (ENS) to the central nervous system (CNS). However, it remains unclear how this process is regulated at a molecular level. Here we show that δ-secretase (asparagine endopeptidase, AEP) cleaves both α-Syn at N103 and Tau at N368, and mediates their fibrillization and retrograde propagation from the gut to the brain, triggering nigra dopaminergic neuronal loss associated with Lewy bodies and motor dysfunction. α-Syn N103 and Tau N368 robustly interact with each other and are highly elevated in PD patients' gut and brain. Chronic oral administration of the neurotoxin rotenone induces AEP activation and α-Syn N103/Tau N368 complex formation in the gut, eliciting constipation and dopaminergic neuronal death in an AEP-dependent manner. Preformed fibrils (PFFs) of α-Syn N103/Tau N368 are more neurotoxic and compact, and aggregate more quickly along the vagus nerve than their FL/FL counterparts or the individual fragments' fibrils. Colonic injection of PFFs induces PD pathologies, motor dysfunctions, and cognitive impairments. Thus, δ-secretase plays a crucial role in initiating PD pathology progression from the ENS to the CNS.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/etiología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/enzimología , Tronco Encefálico , Línea Celular , Células Cultivadas , Colon , Tracto Gastrointestinal/enzimología , Humanos , Ratones , Ovillos Neurofibrilares , Enfermedad de Parkinson/patología , Fosforilación , Ratas , Rotenona/toxicidad , Sinucleinopatías/etiología , Nervio Vago , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/química , Proteínas tau/administración & dosificación , Proteínas tau/química
12.
Acta Neuropathol Commun ; 7(1): 72, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31064413

RESUMEN

Tau is a microtubule stabilizing protein that forms aggregates in Alzheimer's disease (AD). Tau derived from AD patients' brains induces tau aggregation in a prion-like manner when injected into susceptible mouse models.Here we investigated whether cerebrospinal fluid (CSF) collected from patients diagnosed with probable AD or mild cognitive impairment (MCI) likely due to AD harbors a prion-like tau seeding potential. CSF was injected intrahippocampally into young P301S tau transgenic mice. CSF obtained from AD or MCI patients increased hippocampal tau hyperphosphorylation and tau tangle formation in these mice at 4 months post-seeding. Tau pathology was also accentuated in the contralateral hippocampus, and in anterior and posterior directions, indicative of spreading.We provide first evidence for in vivo prion-like properties of AD patients' CSF, accelerating tau pathology in susceptible tau transgenic mice. This demonstrates that biologically active tau seeds reach the CSF compartment in AD. Further studies may help to evaluate strain specific properties of CSF derived tau bioseeds, and to assess their diagnostic potential.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Hipocampo/patología , Agregación Patológica de Proteínas/patología , Proteínas tau/administración & dosificación , Anciano , Anciano de 80 o más Años , Animales , Disfunción Cognitiva/líquido cefalorraquídeo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Agregación Patológica de Proteínas/metabolismo
13.
Behav Brain Res ; 359: 127-134, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30367970

RESUMEN

In order to contribute to a better knowledge on the relationship between amyloid and tau pathology, and electroencephalography (EEG) disturbances, the aim of this study was to evaluate the effects of injection of beta amyloid Abeta(1-42) peptide, tau (a recombinant AAV (Adeno-Associated Virus) containing the human transgene tau with the P301 L mutation on rats and the combination of both, on the power of brain's rhythm (delta, theta, alpha, beta and gamma waves) during the different sleep/wake states of animals by EEG recording. Currently, no preclinical studies explore the effect of the tau pathology on EEG. The experimentations were performed 3 weeks and 3 months post injections. Beta amyloid deposits and hyperphosphorylated Tau are observed by immunohistofluorescence, only in the hippocampus. Furthermore, using a radial arm water maze, the main effect was observed on working memory which was significantly impaired in Abeta-Tau group only 3 months post injections. However, on EEG, as early as the 3rd week, an overall decrease of the EEG bands power was observed in the treated groups, particularly the theta waves during the rapid eye movement (REM) sleep. Beta amyloid was mainly involved in these perturbations. Obviously, EEG seems to be an interesting tool in the early diagnostic of amyloid and tau pathologies, with a good sensitivity and the possibility to perform a follow up during a large period.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/fisiopatología , Electroencefalografía , Fragmentos de Péptidos/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/administración & dosificación , Animales , Dermatoglifia del ADN , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Himecromona , Masculino , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/fisiología , Fragmentos de Péptidos/administración & dosificación , Fosforilación , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sueño REM/fisiología , Proteínas tau/administración & dosificación , Proteínas tau/genética
14.
J Alzheimers Dis ; 63(2): 725-740, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29660939

RESUMEN

Trace eyeblink conditioning is a hippocampus-dependent associative learning paradigm which is impaired in patients with Alzheimer's disease (AD) and animal AD models. Learning in this paradigm accompanies changes in oscillatory activity in forebrain regions, some of which are loci of pathogenic changes in prodromal AD stages. These observations motivated us to examine how cortical event-related potentials (ERPs) during this paradigm are affected by two features of the asymptomatic, AD-related brain abnormality, entorhinal tau accumulation and mild cholinergic deficit. Adult rats received viral overexpression of P301L mutant human tau in the entorhinal cortex, low-dose scopolamine treatment, or both. Electroencephalograms were recorded with epidural electrodes on the surface of the frontal, parietal, and temporal cortices during differential and reversal trace eyeblink conditioning. All rats developed conditioned responses to one of two stimuli (auditory or visual) paired with mild eyelid shock (CS+), but not to the other stimulus presented alone (CS-). They were also able to adjust the response when the stimulus contingency was reversed. With learning, the amplitude of several ERP components in the frontal and temporal cortices came to differentiate the CS+ from CS-. Scopolamine affected the learning-related change in temporal P2 and other learning-unrelated components in three regions. Entorhinal tau overexpression primary affected the amplitude of temporal visual ERPs and learning-unrelated frontal and temporal auditory ERP components. The double manipulation only affected two components of temporal auditory ERPs. Thus, cortical ERPs during differential associative learning are sensitive to asymptomatic brain abnormality associated with AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Aprendizaje por Asociación/fisiología , Encéfalo/fisiopatología , Condicionamiento Palpebral/fisiología , Potenciales Evocados , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/patología , Animales , Percepción Auditiva/fisiología , Encéfalo/patología , Diagnóstico por Computador , Electroencefalografía , Electrochoque , Potenciales Evocados/fisiología , Masculino , Mutación , Ratas Long-Evans , Escopolamina , Máquina de Vectores de Soporte , Percepción Visual/fisiología , Proteínas tau/administración & dosificación , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Alzheimers Res Ther ; 10(1): 13, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386065

RESUMEN

BACKGROUND: Accumulating evidence indicates that Tau pathology can spread from neuron to neuron by intake and coaggregation of the hyperphosphorylated Tau (p-Tau) seeds with the host neuron protein. Thus, clearance of Tau seeds by immunization with Tau antibodies could provide a potential therapeutic opportunity to block the spread of the pathology in Alzheimer's disease (AD) and other tauopathies. We report prevention of the seeding and spread of tau pathology with mouse monoclonal antibody 43D against the N-terminal projection domain of Tau (Tau 6-18) in triple-transgenic AD (3 × Tg-AD) mice. METHODS: Female 11- to 12-month-old 3 × Tg-AD mice were intravenously immunized weekly for 6 weeks with 15 µg/injection of mouse monoclonal antibody 43D or with mouse immunoglobulin G as a control. AD p-Tau isolated from a frozen autopsied AD brain was unilaterally injected into the right hippocampus on the day of the second dose of immunization. Tau pathology and its effect on Aß pathology were assessed by immunohistochemical staining. RESULTS: We found that the injection of AD p-Tau into the hippocampus of 11- to 12-month-old 3 × Tg-AD mice time-dependently induced Tau aggregation in the hippocampus and promoted the spread of Tau pathology to the contralateral hippocampus. Tau pathology was observed as early as 6 weeks after AD p-Tau injection. Tau pathology templated by AD p-Tau was thioflavin-S-positive and was about two-fold greater than that seen in naive 18-month-old 3 × Tg-AD mice; Tau pathology in the latter was thioflavin-S-negative. Immunization with Tau antibody 43D dramatically blocked AD p-Tau seeding in the ipsilateral hippocampus and inhibited its propagation to the contralateral side in 3 × Tg-AD mice. Furthermore, AD p-Tau injection enhanced the amyloid plaque load in the ipsilateral side, and immunization with 43D showed a tendency to attenuate it. CONCLUSIONS: These findings indicate that AD p-Tau-injected 3 × Tg-AD mice represent a practical model to study the seeding and spread of Tau pathology, their effect on Aß pathology, and the effect of Tau immunotherapy on both Tau and Aß pathologies. Immunization with Tau antibody 43D to Tau 6-18 can prevent the seeding and spread of Tau pathology, making it a potential therapeutic treatment for AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Anticuerpos Monoclonales/administración & dosificación , Encéfalo/patología , Inmunización Pasiva , Proteínas tau/inmunología , Aciltransferasas/genética , Aciltransferasas/metabolismo , Administración Intravenosa , Enfermedad de Alzheimer/inmunología , Animales , Encéfalo/inmunología , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Placa Amiloide/inmunología , Placa Amiloide/patología , Placa Amiloide/prevención & control , Presenilina-1/genética , Presenilina-1/metabolismo , Distribución Aleatoria , Proteínas tau/administración & dosificación , Proteínas tau/genética
16.
Methods Cell Biol ; 141: 307-322, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28882310

RESUMEN

The progressive spread of pathological brain lesions containing aggregated tau protein is a hallmark of Alzheimer's disease and other neurodegenerative diseases. In AD, this process follows a distinct pattern along neuronal connections from the entorhinal cortex to hippocampal areas and further on through the limbic system. In other tauopathies, the spread of tau appears less hierarchical throughout the brain, and also nonpathological tau is reported to cross-synaptic connections in the brain. To be able to study the process of cell-to-cell transport of tau and the associated neurotoxicity in the brain in vivo, adeno-associated virus-mediated expression of tau can be used to express different forms of tau in distinct brain areas in rodent models. As an example, we describe how the expression of FTD-mutant human tauP301L in the entorhinal cortex of wild-type mice can be used to study the propagation of tau to connected neurons and to determine pathological consequences such as tau hyperphosphorylation, misfolding, and gliosis. The approach described can easily be translated to study other aggregating and/or propagating proteins in the brain such as synuclein, Abeta, or SOD1.


Asunto(s)
Encéfalo/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Neuronas/patología , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Fosforilación , Tauopatías/metabolismo , Proteínas tau/administración & dosificación , Proteínas tau/genética
17.
J Alzheimers Dis ; 55(1): 411-419, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27662303

RESUMEN

Tauopathies are a hallmark of many neurodegenerative diseases, including Alzheimer's disease and traumatic brain injuries. It has been demonstrated that amyloid-beta peptides, alpha-synuclein, and prion proteins cross the blood-brain barrier (BBB), contributing to their abilities to induce disease. Very little is known about whether tau proteins can cross the BBB. Here we systematically characterized several key forms of tau proteins to cross the BBB, including Tau-441 (2N4R), Tau-410 (2N3R), truncated tau 151-391 (0N4R), and truncated tau 121-227. All of these tau proteins crossed the BBB readily and bidirectonally; however, only Tau-410 had a saturable component to its influx. The tau proteins also entered the blood after their injection into the brain, with Tau 121-227 having the slowest exit from brain. The tau proteins varied in regards to their enzymatic stability in brain and blood and in their peripheral pharmacokinetics. These results show that blood-borne tau proteins could contribute to brain tauopathies. The result also suggest that the CNS can contribute to blood levels of tau, raising the possibility that, as suggested for other misfolded proteins, blood levels of tau proteins could be used as a biomarker of CNS disease.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas tau/metabolismo , Animales , Escherichia coli , Humanos , Radioisótopos de Yodo , Cinética , Masculino , Ratones , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Análisis de Regresión , Proteínas tau/administración & dosificación
18.
J Neurotrauma ; 33(22): 2034-2043, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-26729399

RESUMEN

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


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Proteínas tau/biosíntesis , Proteínas tau/toxicidad , Animales , Disfunción Cognitiva/inducido químicamente , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Técnicas Estereotáxicas/instrumentación , Proteínas tau/administración & dosificación
19.
Exp Neurol ; 248: 451-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23876516

RESUMEN

The recent studies of others and of us showing robust efficacy of anti-tangle immunotherapy, directed against phosphorylated (phos)-tau protein, may pave the way to clinical trials of phos-tau immunotherapy in Alzheimer's-disease and other tauopathies. At this stage addressing the safety of the phos-tau-immunotherapy is highly needed, particularly since we have previously shown the neurotoxic potential of tau-immunotherapy, specifically of full-length unphosphorylated-tau vaccine under a CNS-proinflammatory milieu [induced by emulsification in complete-Freund's-adjuvant (CFA) and pertussis-toxin (PT)] in young wild-type (WT)-mice. The aim of our current study was to address safety aspects of the phos-tau-immunotherapy in both neurofibrillary-tangle (NFT)-mice as well as in WT-mice, under challenging conditions of repeated immunizations with phos-tau peptides under a CNS-proinflammatory milieu. NFT- and WT-mice were repeatedly immunized (7 injections in adult-, 4 in aged-mice) with phos-tau peptides emulsified in CFA-PT. A paralytic disease was evident in the phos-tau-immunized adult NFT-mice, developing progressively to 26.7% with the number of injections. Interestingly, the WT-mice were even more prone to develop neuroinflammation following phos-tau immunization, affecting 75% of the immunized mice. Aged mice were less prone to neuroinflammatory manifestations. Anti-phos-tau antibodies, detected in the serum of immunized mice, partially correlated with the neuroinflammation in WT-mice. This points that repeated phos-tau immunizations in the frame of a proinflammatory milieu may be encephalitogenic to tangle-mice, and more robustly to WT-mice, indicating that - under certain conditions - the safety of phos-tau immunotherapy is questionable.


Asunto(s)
Encefalitis/etiología , Inmunización/efectos adversos , Inmunoterapia/efectos adversos , Tauopatías/etiología , Proteínas tau/administración & dosificación , Proteínas tau/inmunología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Fosforilación , Tauopatías/inmunología , Tauopatías/metabolismo , Proteínas tau/metabolismo
20.
Neurobiol Aging ; 31(12): 2091-102, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19155101

RESUMEN

Microtubule-associated protein tau gene transfer to the substantia nigra of rats using the adeno-associated virus (AAV) vector previously led to neuropathology and neurodegeneration in young rats. In this study, we compared equal tau gene transfer in either 3 or 20-month-old rats, in order to test the hypothesis that late middle-aged rats are more susceptible to neurodegeneration. Two intervals and two vector doses of the tau vector probed for age-related differences in the initial sensitivity to low-level tau expression. Gene transfer efficiency was similar for both ages, but the tau vector caused more dopaminergic cell loss and a greater behavioral deficit in aged rats at specific doses and time points. Tau gene transfer caused microgliosis relative to the control vector, and to a greater extent in aged rats. The maximal microglial response occurred at 2 weeks preceding the peak dopaminergic cell loss by 8 weeks. The cellular and behavioral outcomes were more severe in the aged rats, validating the model for studies of age-related diseases.


Asunto(s)
Envejecimiento/patología , Senescencia Celular , Técnicas de Transferencia de Gen , Gliosis/patología , Microglía/patología , Enfermedades Neurodegenerativas/patología , Proteínas tau/administración & dosificación , Proteínas tau/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Senescencia Celular/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Gliosis/genética , Gliosis/metabolismo , Masculino , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Proteínas tau/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA