Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(11)2023 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-37298211

RESUMEN

The accumulation of tau is a hallmark of several neurodegenerative diseases and is associated with neuronal hypoactivity and presynaptic dysfunction. Oral administration of the adenosine A1 receptor antagonist rolofylline (KW-3902) has previously been shown to reverse spatial memory deficits and to normalize the basic synaptic transmission in a mouse line expressing full-length pro-aggregant tau (TauΔK) at low levels, with late onset of disease. However, the efficacy of treatment remained to be explored for cases of more aggressive tauopathy. Using a combination of behavioral assays, imaging with several PET-tracers, and analysis of brain tissue, we compared the curative reversal of tau pathology by blocking adenosine A1 receptors in three mouse models expressing different types and levels of tau and tau mutants. We show through positron emission tomography using the tracer [18F]CPFPX (a selective A1 receptor ligand) that intravenous injection of rolofylline effectively blocks A1 receptors in the brain. Moreover, when administered to TauΔK mice, rolofylline can reverse tau pathology and synaptic decay. The beneficial effects are also observed in a line with more aggressive tau pathology, expressing the amyloidogenic repeat domain of tau (TauRDΔK) with higher aggregation propensity. Both models develop a progressive tau pathology with missorting, phosphorylation, accumulation of tau, loss of synapses, and cognitive decline. TauRDΔK causes pronounced neurofibrillary tangle assembly concomitant with neuronal death, whereas TauΔK accumulates only to tau pretangles without overt neuronal loss. A third model tested, the rTg4510 line, has a high expression of mutant TauP301L and hence a very aggressive phenotype starting at ~3 months of age. This line failed to reverse pathology upon rolofylline treatment, consistent with a higher accumulation of tau-specific PET tracers and inflammation. In conclusion, blocking adenosine A1 receptors by rolofylline can reverse pathology if the pathological potential of tau remains below a threshold value that depends on concentration and aggregation propensity.


Asunto(s)
Receptor de Adenosina A1 , Tauopatías , Ratones , Animales , Ratones Transgénicos , Receptor de Adenosina A1/genética , Receptor de Adenosina A1/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Hipocampo/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Cognición , Modelos Animales de Enfermedad
2.
Mol Neurodegener ; 14(1): 13, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30917861

RESUMEN

BACKGROUND: Aggregation of tau proteins is a distinct hallmark of tauopathies and has been a focus of research and clinical trials for Alzheimer's Disease. Recent reports have pointed towards a toxic effect of soluble or oligomeric tau in the spreading of tau pathology in Alzheimer's disease. Here we investigated the effects of expressing human tau repeat domain (tauRD) with pro- or anti-aggregant mutations in regulatable transgenic mouse models of Alzheimer's Disease on the functional neuronal networks and the structural connectivity strength. METHODS: Pro-aggregant and anti-aggregant mice were studied when their mutant tauRD was switched on for 12 months to reach the stage where pro-aggregant mice show cognitive impairment, whereas anti-aggregant mice remained cognitively normal. Then, mutant tauRD was switched off by doxycycline treatment for 8 weeks so that soluble transgenic tau disappeared and cognition recovered in the pro-aggregant mice, although some aggregates remained. At these two time points, at baseline after 12 months of mutant tau expression and after 8 weeks of doxycycline treatment, resting state fMRI and diffusion MRI were used to determine functional neuronal networks and fiber connectivities. Results of the transgenic mice were compared with wildtype littermates. RESULTS: Functional connectivity was strongly reduced in transgenic animals during mutant tauRD expression, in relation to WT mice. Interestingly, transgenic mice with the non-aggregant tau mutant showed identical functional deficits as the pro-aggregant mice, even though in this case there was no cognitive decline by behavioral testing. Upon 8 weeks doxycycline treatment and transgene switch-off, functional connectivity in both transgenic groups presented complete normalization of functional connectivity strength, equivalent to the situation in WT littermates. Structural connectivity was found only marginally sensitive to mutant tau expression (both pro- and anti-aggregant tauRD) and by doxycycline treatment. CONCLUSIONS: Our in vivo investigations unravel for the first time a strong reduction of functional neuronal networks by the presence of increased soluble rather than fibrillary tau, independent of its intrinsic propensity of aggregation, which is reversible by switching tau off. Our functional MRI study thus is an unexpected in vivo validation of a novel property of tau, while previous results pointed to a role of aggregation propensity for a pathological state by histopathology and cognitive decline. Our results present further evidence for early tauopathy biomarkers or a potential early stage drug target by functional networks analysis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Modelos Animales de Enfermedad , Memoria/fisiología , Proteínas tau/metabolismo , Animales , Cognición/fisiología , Hipocampo/metabolismo , Ratones , Red Nerviosa/fisiopatología , Neuronas/metabolismo , Sinapsis/metabolismo , Tauopatías/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(41): 11597-11602, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671637

RESUMEN

Accumulation of Tau is a characteristic hallmark of several neurodegenerative diseases but the mode of toxic action of Tau is poorly understood. Here, we show that the Tau protein is toxic due to its aggregation propensity, whereas phosphorylation and/or missorting is not sufficient to cause neuronal dysfunction. Aggregate-prone Tau accumulates, when expressed in vitro at near-endogenous levels, in axons as spindle-shaped grains. These axonal grains contain Tau that is folded in a pathological (MC-1) conformation. Proaggregant Tau induces a reduction of neuronal ATP, concomitant with loss of dendritic spines. Counterintuitively, axonal grains of Tau are not targeted for degradation and do not induce a molecular stress response. Proaggregant Tau causes neuronal and astrocytic hypoactivity and presynaptic dysfunction instead. Here, we show that the adenosine A1 receptor antagonist rolofylline (KW-3902) is alleviating the presynaptic dysfunction and restores neuronal activity as well as dendritic spine levels in vitro. Oral administration of rolofylline for 2-wk to 14-mo-old proaggregant Tau transgenic mice restores the spatial memory deficits and normalizes the basic synaptic transmission. These findings make rolofylline an interesting candidate to combat the hypometabolism and neuronal dysfunction associated with Tau-induced neurodegenerative diseases.


Asunto(s)
Antagonistas del Receptor de Adenosina A1/farmacología , Axones/metabolismo , Eliminación de Secuencia , Xantinas/farmacología , Proteínas tau/genética , Adenosina Trifosfato/metabolismo , Animales , Espinas Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Agregado de Proteínas , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Tauopatías/tratamiento farmacológico , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología
4.
EMBO Rep ; 17(4): 552-69, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26931569

RESUMEN

We report on a novel transgenic mouse model expressing human full-length Tau with the Tau mutation A152T (hTau(AT)), a risk factor for FTD-spectrum disorders including PSP and CBD Brain neurons reveal pathological Tau conformation, hyperphosphorylation, mis-sorting, aggregation, neuronal degeneration, and progressive loss, most prominently in area CA3 of the hippocampus. The mossy fiber pathway shows enhanced basal synaptic transmission without changes in short- or long-term plasticity. In organotypic hippocampal slices, extracellular glutamate increases early above control levels, followed by a rise in neurotoxicity. These changes are normalized by inhibiting neurotransmitter release or by blocking voltage-gated sodium channels. CA3 neurons show elevated intracellular calcium during rest and after activity induction which is sensitive to NR2B antagonizing drugs, demonstrating a pivotal role of extrasynaptic NMDA receptors. Slices show pronounced epileptiform activity and axonal sprouting of mossy fibers. Excitotoxic neuronal death is ameliorated by ceftriaxone, which stimulates astrocytic glutamate uptake via the transporter EAAT2/GLT1. In summary, hTau(AT) causes excitotoxicity mediated by NR2B-containing NMDA receptors due to enhanced extracellular glutamate.


Asunto(s)
Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Demencia Frontotemporal/fisiopatología , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Calcio/metabolismo , Medios de Cultivo/química , Modelos Animales de Enfermedad , Ácido Glutámico/análisis , Humanos , Ratones , Ratones Transgénicos , Mutación , Plasticidad Neuronal , Neuronas/química , Neuronas/patología , Técnicas de Cultivo de Órganos , Transmisión Sináptica , Proteínas tau/química
5.
Acta Neuropathol Commun ; 4: 17, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26916334

RESUMEN

INTRODUCTION: Mutations of Tau are associated with several neurodegenerative disorders. Recently, the Tau mutation A152T was described as a novel risk factor for frontotemporal dementia spectrum disorders and Alzheimer disease. In vitro Tau-A152T shows a decreased binding to microtubules and a reduced tendency to form abnormal fibers. RESULTS: To study the effects of this mutation we generated a mouse model expressing human full-length Tau with this mutation (hTau40(AT)). At young age (2-3 months) immunohistological analysis reveals pathological Tau conformation and Tau-hyperphosphorylation combined with Tau missorting into the somatodendritic compartment of neurons. With increasing age there is Tau aggregation including co-aggregates of endogenous mouse Tau and exogenous human Tau, accompanied by loss of synapses (especially presynaptic failure) and neurons. From ~10 months onwards the mice show a prominent neuroinflammatory response as judged by activation of microglia and astrocytes. This progressive neuroinflammation becomes visible by in vivo bioluminescence imaging after crossbreeding of hTau40(AT) mice and Gfap-luciferase reporter mice. In contrast to other Tau-transgenic models and Alzheimer disease patients with reduced protein clearance, hTau40(AT) mice show a strong induction of autophagy. Although Tau-hyperphosphorylation and aggregation is also present in spinal cord and motor cortex (due to the Thy1.2 promoter), neuromotor performance is not affected. Deficits in spatial reference memory are manifest at ~16 months and are accompanied by neuronal death. CONCLUSIONS: The hTau40(AT) mice mimic pathological hallmarks of tauopathies including a cognitive phenotype combined with pronounced neuroinflammation visible by bioluminescence. Thus the mice are suitable for mechanistic studies of Tau induced toxicity and in vivo validation of neuroprotective compounds.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Trastornos del Conocimiento/etiología , Encefalitis/etiología , Neumotórax/complicaciones , Neumotórax/genética , Proteínas tau/genética , Factores de Edad , Alanina/genética , Animales , Astrocitos/patología , Astrocitos/ultraestructura , Citocinas/metabolismo , Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Treonina/genética , Proteínas tau/metabolismo
6.
Acta Neuropathol Commun ; 3: 25, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25958115

RESUMEN

INTRODUCTION: Neurofibrillary tangles (NFT) composed of Tau are hallmarks of neurodegeneration in Alzheimer disease. Transgenic mice expressing full-length pro-aggregant human Tau (2N4R Tau-ΔK280, termed Tau(ΔK)) or its repeat domain (TauRD-ΔK280, TauRD(ΔK)) develop a progressive Tau pathology with missorting, phosphorylation, aggregation of Tau, loss of synapses and functional deficits. Whereas TauRD(ΔK) assembles into NFT concomitant with neuronal death, Tau(ΔK) accumulates into Tau pretangles without overt neuronal loss. Both forms cause a comparable cognitive decline (with onset at 10mo and 12mo, respectively), which is rescued upon switch-off of transgene expression. Since methylene blue (MB) is able to inhibit Tau aggregation in vitro, we investigated whether MB can prevent or rescue Tau-induced cognitive impairments in our mouse models. Both types of mice received MB orally using different preventive and therapeutic treatment protocols, initiated either before or after disease onset. The cognitive status of the mice was assessed by behavior tasks (open field, Morris water maze) to determine the most successful conditions for therapeutic intervention. RESULTS: Preventive and therapeutic MB application failed to avert or recover learning and memory deficits of TauRD(ΔK) mice. Similarly, therapeutic MB treatment initiated after onset of cognitive impairments was ineffective in Tau(ΔK) mice. In contrast, preventive MB application starting before onset of functional deficits preserved cognition of Tau(ΔK) mice. Beside improved learning and memory, MB-treated Tau(ΔK) mice showed a strong decrease of insoluble Tau, a reduction of conformationally changed (MC1) and phosphorylated Tau species (AT180, PHF1) as well as an upregulation of protein degradation systems (autophagy and proteasome). This argues for additional pleiotropic effects of MB beyond its properties as Tau aggregation inhibitor. CONCLUSIONS: Our data support the use of Tau aggregation inhibitors as potential drugs for the treatment of AD and other tauopathies and highlights the need for preventive treatment before onset of cognitive impairments.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/prevención & control , Azul de Metileno/farmacología , Tauopatías/tratamiento farmacológico , Proteínas tau/genética , Animales , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Azul de Metileno/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tauopatías/genética , Tauopatías/psicología , Factores de Tiempo , Resultado del Tratamiento , Proteínas tau/metabolismo
7.
Acta Neuropathol Commun ; 3: 23, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25853683

RESUMEN

INTRODUCTION: We used an inducible mouse model expressing the Tau repeat domain with the pro-aggregant mutation ΔK280 to analyze presynaptic Tau pathology in the hippocampus. RESULTS: Expression of pro-aggregant Tau(RDΔ) leads to phosphorylation, aggregation and missorting of Tau in area CA3. To test presynaptic pathophysiology we used electrophysiology in the mossy fiber tract. Synaptic transmission was severely disturbed in pro-aggregant Tau(RDΔ) and Tau-knockout mice. Long-term depression of the mossy fiber tract failed in pro-aggregant Tau(RDΔ) mice. We observed an increase in bouton size, but a decline in numbers and presynaptic markers. Both pre-and postsynaptic structural deficits are preventable by inhibition of Tau(RDΔ) aggregation. Calcium imaging revealed progressive calcium dysregulation in boutons of pro-aggregant Tau(RDΔ) mice. In N2a cells we observed this even in cells without tangle load, whilst in primary hippocampal neurons transient Tau(RDΔ) expression alone caused similar Ca(++) dysregulation. Ultrastructural analysis revealed a severe depletion of synaptic vesicles pool in accordance with synaptic transmission impairments. CONCLUSIONS: We conclude that oligomer formation by Tau(RDΔ) causes pre- and postsynaptic structural deterioration and Ca(++) dysregulation which leads to synaptic plasticity deficits.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/fisiología , Fibras Musgosas del Hipocampo/fisiología , Plasticidad Neuronal/fisiología , Transmisión Sináptica/fisiología , Proteínas tau/metabolismo , Animales , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Electrofisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Noqueados , Proteínas tau/deficiencia , Proteínas tau/genética
8.
FEBS J ; 280(18): 4371-81, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23517246

RESUMEN

Accumulation of amyloidogenic proteins such as Tau is a hallmark of neurodegenerative diseases including Alzheimer disease and fronto-temporal dementias. To link Tau pathology to cognitive impairments and defects in synaptic plasticity, we created four inducible Tau transgenic mouse models with expression of pro- and anti-aggregant variants of either full-length human Tau (hTau40/ΔK280 and hTau40/ΔK280/PP) or the truncated Tau repeat domain (Tau(RD)/ΔK280 and Tau(RD)/ΔK280/PP). Here we review the histopathological features caused by pro-aggregant Tau, and correlate them with behavioral deficits and impairments in synaptic transmission. Both pro-aggregant Tau variants cause Alzheimer-like features, including synapse loss, mis-localization of Tau into the somatodendritic compartment, conformational changes and hyperphosphorylation. However, there is a clear difference in the extent of Tau aggregation and neurotoxicity. While pro-aggregant full-length hTau40/ΔK280 leads to a 'pre-tangle' pathology, the repeat domain Tau(RD)/ΔK280 causes massive formation of neurofibrillary tangles and neuronal loss in the hippocampus. However, both Tau variants cause co-aggregation of human and mouse Tau and similar functional impairments. Thus, earlier Tau pathological stages and not necessarily neurofibrillary tangles are critical for the development of cognitive malfunctions. Most importantly, memory and synapses recover after switching off expression of pro-aggregant Tau. The rescue of functional impairments correlates with the rescue of most Tau pathological changes and most strikingly the recovery of synapses. This implies that tauopathies as such are reversible, provided that amyloidogenic Tau is removed. Therefore, our Tau transgenic mice may serve as model systems for in vivo validation of therapeutic strategies and drug candidates with regard to cognition and synaptic function.


Asunto(s)
Enfermedad de Alzheimer/patología , Hipocampo/patología , Ovillos Neurofibrilares/patología , Sinapsis/patología , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipocampo/metabolismo , Humanos , Memoria , Ratones , Mutación , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Estructura Terciaria de Proteína , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica , Proteínas tau/química , Proteínas tau/metabolismo
9.
Acta Neuropathol ; 123(6): 787-805, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22532069

RESUMEN

Neurofibrillary lesions of abnormal Tau are hallmarks of Alzheimer disease and frontotemporal dementias. Our regulatable (Tet-OFF) mouse models of tauopathy express variants of human full-length Tau in the forebrain (CaMKIIα promoter) either with mutation ΔK280 (pro-aggregant) or ΔK280/I277P/I308P (anti-aggregant). Co-expression of luciferase enables in vivo quantification of gene expression by bioluminescence imaging. Pro-aggregant mice develop synapse loss and Tau-pathology including missorting, phosphorylation and early pretangle formation, whereas anti-aggregant mice do not. We correlated hippocampal Tau pathology with learning/memory performance and synaptic plasticity. Pro-aggregant mice at 16 months of gene expression exhibited severe cognitive deficits in Morris water maze and in passive-avoidance paradigms, whereas anti-aggregant mice were comparable to controls. Cognitive impairment of pro-aggregant mice was accompanied by loss of hippocampal LTP in CA1 and CA3 areas and by a reduction of synaptic proteins and dendritic spines, although no neuronal loss was observed. Remarkably, memory and LTP recovered when pro-aggregant Tau was switched-OFF for ~4 months, Tau phosphorylation and missorting were reversed, and synapses recovered. Moreover, soluble and insoluble pro-aggregant hTau40 disappeared, while insoluble mouse Tau was still present. This study links early Tau pathology without neurofibrillary tangles and neuronal death to cognitive decline and synaptic dysfunction. It demonstrates that Tau-induced impairments are reversible after switching-OFF pro-aggregant Tau. Therefore, our mouse model may mimic an early phase of AD when the hippocampus does not yet suffer from irreversible cell death but cognitive deficits are already striking. It offers potential to evaluate drugs with regard to learning and memory performance.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Hipocampo/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Trastornos del Conocimiento/genética , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/fisiopatología , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Pruebas Neuropsicológicas , Sinapsis/genética , Sinapsis/patología , Tauopatías/metabolismo , Tauopatías/patología , Factores de Tiempo , Proteínas tau/genética
10.
J Mol Neurosci ; 45(3): 432-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21822709

RESUMEN

The accumulation of proteins such as Tau is a hallmark of several neurodegenerative diseases, e.g., frontotemporal dementia (FTD). So far, many mouse models of tauopathies have been generated by the use of mutated or truncated human Tau isoforms in order to enhance the amyloidogenic character of Tau and to mimic pathological processes similar to those in FTD patients. Our inducible mice express the repeat domain of human Tau (Tau(RD)) carrying the FTDP-17 mutation ΔK280 in a "pro-aggregant" and an "anti-aggregant" version. Based on the enhanced tendency of Tau to aggregate, only the "pro-aggregant" Tau(RD) mice develop Tau pathology (hyperphosphorylation, coassembly of human and mouse Tau, synaptic loss, and neuronal degeneration). We have now carried out behavioral and electrophysiological analyses showing that only the pro-aggregant Tau(RD) mice have impaired learning/memory and a distinct loss of LTP. Remarkably, after suppressing the pro-aggregant human Tau(RD), memory and LTP recover, while neuronal loss persists. Aggregates persist as well but change their composition from mixed human/mouse to mouse Tau only. The rescue of cognition and synaptic plasticity is explained by a partial recovery of spine synapses in the hippocampus. These results indicate a tight relationship between the amyloidogenic character of Tau and brain malfunction, and suggest that the cognitive impairment is caused by toxic human Tau(RD) species rather than by mouse Tau aggregates.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Ratones Transgénicos , Tauopatías/fisiopatología , Proteínas tau/metabolismo , Animales , Trastornos del Conocimiento/patología , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Ratones , Mutación , Tauopatías/genética , Tauopatías/patología , Proteínas tau/química , Proteínas tau/genética
11.
J Neurosci ; 31(7): 2511-25, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21325519

RESUMEN

This report describes the behavioral and electrophysiological analysis of regulatable transgenic mice expressing mutant repeat domains of human Tau (Tau(RD)). Mice were generated to express Tau(RD) in two forms, differing in their propensity for ß-structure and thus in their tendency for aggregation ("pro-aggregant" or "anti-aggregant") (Mocanu et al., 2008). Only pro-aggregant mice show pronounced changes typical for Tau pathology in Alzheimer's disease (aggregation, missorting, hyperphosphorylation, synaptic and neuronal loss), indicating that the ß-propensity and hence the ability to aggregate is a key factor in the disease. We now tested the mice with regard to neuromotor parameters, behavior, learning and memory, and synaptic plasticity and correlated this with histological and biochemical parameters in different stages of switching Tau(RD) on or off. The mice are normal in neuromotor tests. However, pro-aggregant Tau(RD) mice are strongly impaired in memory and show pronounced loss of long-term potentiation (LTP), suggesting that Tau aggregation specifically perturbs these brain functions. Remarkably, when the expression of human pro-aggregant Tau(RD) is switched on for ∼ 10 months and off for ∼ 4 months, memory and LTP recover, whereas aggregates decrease moderately and change their composition from mixed human plus mouse Tau to mouse Tau only. Neuronal loss persists, but synapses are partially rescued. This argues that continuous presence of amyloidogenic pro-aggregant Tau(RD) constitutes the main toxic insult for memory and LTP, rather than the aggregates as such.


Asunto(s)
Potenciación a Largo Plazo/genética , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Mutación/genética , Tauopatías , Proteínas tau/genética , Animales , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Fuerza de la Mano/fisiología , Hipocampo/patología , Hipocampo/ultraestructura , Humanos , Técnicas In Vitro , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Pruebas Neuropsicológicas , Estructura Terciaria de Proteína , Percepción Espacial/fisiología , Sinapsis/patología , Sinapsis/ultraestructura , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/fisiopatología , Factores de Tiempo , Proteínas tau/metabolismo
12.
Neurodegener Dis ; 7(1-3): 28-31, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20160454

RESUMEN

BACKGROUND: Aggregates of the tau protein are a hallmark of Alzheimer's and several other neurodegenerative diseases. Various transgenic mouse models have been generated to study the aggregation process. Since wild-type tau is highly soluble and does not aggregate readily, most models make use of tau mutations that occur in human frontotemporal dementias and are more prone to aggregate. These mouse models show neurofibrillary tangles similar to those of Alzheimer's disease. However, since the mice contain both endogenous wild-type mouse tau and exogenous human mutant tau, the relative contribution of these components to the aggregates has been a matter of debate. OBJECTIVE: Using a new set of regulatable transgenic mouse models, we sought to determine whether mouse tau coaggregates with human tau when it is switched on. Furthermore, we asked what type of tau remains in the aggregates after switching off the expression of exogenous tau. METHODS: We generated doxycycline-inducible transgenic mice expressing either full-length human tau or the tau repeat domain (tau(RD)). In addition, both types of human tau derivatives were expressed in a 'proaggregant' form (with the frontotemporal dementia with parkinsonism linked to chromosome 17 mutation DeltaK280), or in an 'antiaggregant' form (with additional proline mutations to block beta-structure and aggregation). RESULTS: The proaggregant tau(RD) mice develop tangles rapidly after induction, the antiaggregant mice do not. Analysis by biochemistry and immunohistology reveals that the tangles contain both exogenous and endogenous mouse tau. After switching off the proaggregant tau(RD), tangles persist for extended periods. However, they are composed entirely of mouse tau. CONCLUSIONS: Mouse tau and exogenous human tau can coaggregate in transgenic models of tauopathy. The aggregates are in dynamic equilibrium with their subunits, so that exogenous tau disappears when its expression is switched off. Once the seeds of aggregation are generated by the foreign tau species, they propagate in a 'prion-like' fashion within the cell even after the foreign tau has disappeared.


Asunto(s)
Modelos Animales de Enfermedad , Prolina/genética , Tauopatías/metabolismo , Proteínas tau/metabolismo , Factores de Edad , Animales , Encéfalo/metabolismo , Encéfalo/patología , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Estructura Terciaria de Proteína/genética , Tauopatías/patología , Proteínas tau/genética
13.
J Clin Invest ; 119(5): 1382-95, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363289

RESUMEN

Our aging society is confronted with a dramatic increase of patients suffering from tauopathies, which include Alzheimer disease and certain frontotemporal dementias. These disorders are characterized by typical neuropathological lesions including hyperphosphorylation and subsequent aggregation of TAU protein and neuronal cell death. Currently, no mechanism-based cures are available. We generated fluorescently labeled TAU transgenic zebrafish, which rapidly recapitulated key pathological features of tauopathies, including phosphorylation and conformational changes of human TAU protein, tangle formation, neuronal and behavioral disturbances, and cell death. Due to their optical transparency and small size, zebrafish larvae are well suited for both in vivo imaging and drug development. TAU-induced neuronal cell death was imaged by time-lapse microscopy in vivo. Furthermore, we used this zebrafish model to identify compounds targeting the TAU kinase glycogen synthase kinase 3beta (GSK3beta). We identified a newly developed highly active GSK3beta inhibitor, AR-534, by rational drug design. AR-534 reduced TAU phosphorylation in TAU transgenic zebrafish. This transgenic zebrafish model may become a valuable tool for further studies of the neuropathology of dementia.


Asunto(s)
Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Neuronas/patología , Tauopatías/tratamiento farmacológico , Tauopatías/patología , Pez Cebra , Animales , Animales Modificados Genéticamente , Muerte Celular , Diseño de Fármacos , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Reacción de Fuga , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Larva/anatomía & histología , Larva/efectos de los fármacos , Larva/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Modelos Moleculares , Estructura Molecular , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Conformación Proteica , Alineación de Secuencia , Médula Espinal/metabolismo , Médula Espinal/patología , Sinaptotagminas/metabolismo , Tauopatías/metabolismo , Pez Cebra/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...