Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 142(3): 387-97, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20655099

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid-beta (Abeta) and tau deposition in brain. It has emerged that Abeta toxicity is tau dependent, although mechanistically this link remains unclear. Here, we show that tau, known as axonal protein, has a dendritic function in postsynaptic targeting of the Src kinase Fyn, a substrate of which is the NMDA receptor (NR). Missorting of tau in transgenic mice expressing truncated tau (Deltatau) and absence of tau in tau(-/-) mice both disrupt postsynaptic targeting of Fyn. This uncouples NR-mediated excitotoxicity and hence mitigates Abeta toxicity. Deltatau expression and tau deficiency prevent memory deficits and improve survival in Abeta-forming APP23 mice, a model of AD. These deficits are also fully rescued with a peptide that uncouples the Fyn-mediated interaction of NR and PSD-95 in vivo. Our findings suggest that this dendritic role of tau confers Abeta toxicity at the postsynapse with direct implications for pathogenesis and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Dendritas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Encéfalo/patología , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Proteínas tau/genética
2.
Acta Neuropathol ; 144(4): 637-650, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35780436

RESUMEN

In Alzheimer's disease (AD), where amyloid-ß (Aß) and tau deposits in the brain, hyperexcitation of neuronal networks is an underlying disease mechanism, but its cause remains unclear. Here, we used the Collaborative Cross (CC) forward genetics mouse platform to identify modifier genes of neuronal hyperexcitation. We found LAMP5 as a novel regulator of hyperexcitation in mice, critical for the survival of distinct interneuron populations. Interestingly, synaptic LAMP5 was lost in AD brains and LAMP5 interneurons degenerated in different AD mouse models. Genetic reduction of LAMP5 augmented functional deficits and neuronal network hypersynchronicity in both Aß- and tau-driven AD mouse models. To this end, our work defines the first specific function of LAMP5 interneurons in neuronal network hyperexcitation in AD and dementia with tau pathology.


Asunto(s)
Enfermedad de Alzheimer , Proteínas de Membrana de los Lisosomas/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/fisiología , Animales , Modelos Animales de Enfermedad , Interneuronas/patología , Ratones , Ratones Transgénicos , Neuronas/patología , Proteínas tau/genética
3.
Biochem J ; 478(7): 1471-1484, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33769438

RESUMEN

Tau pathology initiates in defined brain regions and is known to spread along neuronal connections as symptoms progress in Alzheimer's disease (AD) and other tauopathies. This spread requires the release of tau from donor cells, but the underlying molecular mechanisms remained unknown. Here, we established the interactome of the C-terminal tail region of tau and identified syntaxin 8 (STX8) as a mediator of tau release from cells. Similarly, we showed the syntaxin 6 (STX6), part of the same SNARE family as STX8 also facilitated tau release. STX6 was previously genetically linked to progressive supranuclear palsy (PSP), a tauopathy. Finally, we demonstrated that the transmembrane domain of STX6 is required and sufficient to mediate tau secretion. The differential role of STX6 and STX8 in alternative secretory pathways suggests the association of tau with different secretory processes. Taken together, both syntaxins, STX6 and STX8, may contribute to AD and PSP pathogenesis by mediating release of tau from cells and facilitating pathology spreading.


Asunto(s)
Enfermedad de Alzheimer/patología , Dominios y Motivos de Interacción de Proteínas , Proteínas Qa-SNARE/metabolismo , Vías Secretoras , Tauopatías/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Unión Proteica , Proteínas Qa-SNARE/genética , Tauopatías/genética , Tauopatías/metabolismo , Proteínas tau/genética
4.
J Biol Chem ; 294(38): 14149-14162, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31366728

RESUMEN

The microtubule-associated protein tau undergoes aberrant modification resulting in insoluble brain deposits in various neurodegenerative diseases, including frontotemporal dementia (FTD), progressive supranuclear palsy, and corticobasal degeneration. Tau aggregates can form in different cell types of the central nervous system (CNS) but are most prevalent in neurons. We have previously recapitulated aspects of human FTD in mouse models by overexpressing mutant human tau in CNS neurons, including a P301S tau variant in TAU58/2 mice, characterized by early-onset and progressive behavioral deficits and FTD-like neuropathology. The molecular mechanisms underlying the functional deficits of TAU58/2 mice remain mostly elusive. Here, we employed functional genomics (i.e. RNAseq) to determine differentially expressed genes in young and aged TAU58/2 mice to identify alterations in cellular processes that may contribute to neuropathy. We identified genes in cortical brain samples differentially regulated between young and old TAU58/2 mice relative to nontransgenic littermates and by comparative analysis with a dataset of CNS cell type-specific genes expressed in nontransgenic mice. Most differentially-regulated genes had known or putative roles in neurons and included presynaptic and excitatory genes. Specifically, we observed changes in presynaptic factors, glutamatergic signaling, and protein scaffolding. Moreover, in the aged mice, expression levels of several genes whose expression was annotated to occur in other brain cell types were altered. Immunoblotting and immunostaining of brain samples from the TAU58/2 mice confirmed altered expression and localization of identified and network-linked proteins. Our results have revealed genes dysregulated by progressive tau accumulation in an FTD mouse model.


Asunto(s)
Tauopatías/genética , Tauopatías/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Demencia Frontotemporal/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Análisis de Secuencia de ARN/métodos , Tauopatías/fisiopatología , Proteínas tau/metabolismo
5.
Am J Pathol ; 188(6): 1447-1456, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29577934

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing and fatal disease characterized by muscular atrophy because of loss of upper and lower motor neurons. Histopathologically, most patients with ALS have abnormal cytoplasmic accumulation and aggregation of the nuclear RNA-regulating protein TAR DNA-binding protein 43 (TDP-43). Pathogenic mutations in the TARDBP gene that encode TDP-43 have been identified in familial ALS. We have previously reported transgenic mice with neuronal expression of human TDP-43 carrying the pathogenic A315T mutation (iTDP-43A315T mice), presenting with early-onset motor deficits in adolescent animals. Here, we analyzed aged iTDP-43A315T mice, focusing on the spatiotemporal profile and progression of neurodegeneration in upper and lower motor neurons. Magnetic resonance imaging and histologic analysis revealed a differential loss of upper motor neurons in a hierarchical order as iTDP-43A315T mice aged. Furthermore, we report progressive gait problems, profound motor deficits, and muscle atrophy in aged iTDP-43A315T mice. Despite these deficits and TDP-43 pathologic disorders in lower motor neurons, stereological analysis did not show cell loss in spinal cords. Taken together, neuronal populations in aging iTDP-43A315T mice show differential susceptibility to the expression of human TDP-43A315T.


Asunto(s)
Sistema Nervioso Central/patología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Trastornos Motores/patología , Atrofia Muscular/patología , Enfermedades Neurodegenerativas/patología , Envejecimiento , Animales , Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Motores/genética , Trastornos Motores/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Análisis Espacio-Temporal
6.
Brain ; 141(2): 348-356, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29272336

RESUMEN

Faciobrachial dystonic seizures and limbic encephalitis closely associate with antibodies to leucine-rich glioma-inactivated 1 (LGI1). Here, we describe 103 consecutive patients with faciobrachial dystonic seizures and LGI1 antibodies to understand clinical, therapeutic and serological differences between those with and without cognitive impairment, and to determine whether cessation of faciobrachial dystonic seizures can prevent cognitive impairment. The 22/103 patients without cognitive impairment typically had normal brain MRI, EEGs and serum sodium levels (P < 0.0001). Overall, cessation of faciobrachial dystonic seizures with antiepileptic drugs alone occurred in only 9/89 (10%) patients. By contrast, 51% showed cessation of faciobrachial dystonic seizures 30 days after addition of immunotherapy (P < 0.0001), with earlier cessation in cognitively normal patients (P = 0.038). Indeed, expedited immunotherapy (P = 0.031) and normal cognition (P = 0.0014) also predicted reduced disability at 24 months. Furthermore, of 80 patients with faciobrachial dystonic seizures as their initial feature, 56% developed cognitive impairment after 90 days of active faciobrachial dystonic seizures. Whereas only one patient developed cognitive impairment after cessation of faciobrachial dystonic seizures (P < 0.0001). All patients had IgG4-LGI1 antibodies, but those with cognitive impairment had higher proportions of complement-fixing IgG1 antibodies (P = 0.03). Both subclasses caused LGI1-ADAM22 complex internalization, a potential non-inflammatory epileptogenic mechanism. In summary, faciobrachial dystonic seizures show striking time-sensitive responses to immunotherapy, and their cessation can prevent the development of cognitive impairment.awx323media15681705685001.


Asunto(s)
Inmunoterapia/métodos , Encefalitis Límbica/complicaciones , Convulsiones/etiología , Convulsiones/terapia , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Anticuerpos/sangre , Anticuerpos/metabolismo , Anticonvulsivantes/uso terapéutico , Trastornos del Conocimiento/etiología , Personas con Discapacidad , Femenino , Citometría de Flujo , Estudios de Seguimiento , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Encefalitis Límbica/sangre , Encefalitis Límbica/terapia , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas/fisiología , Proteínas/inmunología , Estudios Retrospectivos , Encuestas y Cuestionarios , Transfección , Adulto Joven
7.
Acta Neuropathol ; 130(5): 661-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26437864

RESUMEN

The nuclear transactive response DNA-binding protein 43 (TDP-43) undergoes relocalization to the cytoplasm with formation of cytoplasmic deposits in neurons in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Pathogenic mutations in the TDP-43-encoding TARDBP gene in familial ALS as well as non-mutant human TDP-43 have been utilized to model FTD/ALS in cell culture and animals, including mice. Here, we report novel A315T mutant TDP-43 transgenic mice, iTDP-43(A315T), with controlled neuronal over-expression. Constitutive expression of human TDP-43(A315T) resulted in pronounced early-onset and progressive neurodegeneration, which was associated with compromised motor performance, spatial memory and disinhibition. Muscle atrophy resulted in reduced grip strength. Cortical degeneration presented with pronounced astrocyte activation. Using differential protein extraction from iTDP-43(A315T) brains, we found cytoplasmic localization, fragmentation, phosphorylation and ubiquitination and insolubility of TDP-43. Surprisingly, suppression of human TDP-43(A315T) expression in mice with overt neurodegeneration for only 1 week was sufficient to significantly improve motor and behavioral deficits, and reduce astrogliosis. Our data suggest that functional deficits in iTDP-43(A315T) mice are at least in part a direct and transient effect of the presence of TDP-43(A315T). Furthermore, it illustrates the compensatory capacity of compromised neurons once transgenic TDP-43 is removed, with implications for future treatments.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Degeneración Lobar Frontotemporal/fisiopatología , Mutación , Recuperación de la Función/fisiología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Astrocitos/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Doxiciclina , Femenino , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Gliosis/patología , Gliosis/fisiopatología , Fuerza de la Mano/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/patología , Memoria Espacial/fisiología
8.
J Neural Transm (Vienna) ; 121(5): 543-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24337667

RESUMEN

Ischemic stroke is a leading cause of death. It has previously been shown that blocking activation of extracellular signal-regulated kinase (ERK) with the MEK inhibitor U0126 mitigates brain damage in rodent models of ischemic stroke. Here we show that the newer MEK inhibitor PD184161 reduces cell death and altered gene expression in cultured neurons and mice undergoing excitotoxicity, and has similar protective effects in a mouse model of stroke. This further supports ERK inhibition as a potential treatment for stroke.


Asunto(s)
Compuestos de Anilina/farmacología , Benzamidas/farmacología , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Ácido Glutámico/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratones Endogámicos C57BL , Neuronas/fisiología , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología
9.
Proc Natl Acad Sci U S A ; 105(41): 15997-6002, 2008 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-18832465

RESUMEN

Frontotemporal dementia (FTD) is characterized by cognitive and behavioral changes and, in a significant subset of patients, Parkinsonism. Histopathologically, FTD frequently presents with tau-containing lesions, which in familial cases result from mutations in the MAPT gene encoding tau. Here we present a novel transgenic mouse strain (K3) that expresses human tau carrying the FTD mutation K369I. K3 mice develop a progressive histopathology that is reminiscent of that in human FTD with the K369I mutation. In addition, K3 mice show early-onset memory impairment and amyotrophy in the absence of overt neurodegeneration. Different from our previously generated tau transgenic strains, the K3 mice express the transgene in the substantia nigra (SN) and show an early-onset motor phenotype that reproduces Parkinsonism with tremor, bradykinesia, abnormal gait, and postural instability. Interestingly, motor performance of young, but not old, K3 mice improves upon L-dopa treatment, which bears similarities to Parkinsonism in FTD. The early-onset symptoms in the K3 mice are mechanistically related to selectively impaired anterograde axonal transport of distinct cargos, which precedes the loss of dopaminergic SN neurons that occurs in aged mice. The impaired axonal transport in SN neurons affects, among others, vesicles containing the dopamine-synthesizing enzyme tyrosine hydroxylase. Distinct modes of transport are also impaired in sciatic nerves, which may explain amyotrophy. Together, the K3 mice are a unique model of FTD-associated Parkinsonism, with pathomechanistic implications for the human pathologic process.


Asunto(s)
Transporte Axonal , Demencia/fisiopatología , Modelos Animales de Enfermedad , Trastornos Parkinsonianos/fisiopatología , Animales , Demencia/patología , Lóbulo Frontal , Humanos , Levodopa/farmacología , Ratones , Ratones Transgénicos , Trastornos de la Destreza Motora/genética , Mutación Missense , Trastornos Parkinsonianos/genética , Fenotipo , Nervio Ciático , Sustancia Negra/patología , Lóbulo Temporal , Proteínas tau/genética
10.
J Neural Transm (Vienna) ; 116(10): 1243-51, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19693433

RESUMEN

Frontotemporal lobar degeneration (FTLD) is a common cause of presenile dementia characterised by behavioural and language disturbances. Pick's disease (PiD) is a subtype of FTLD, which presents with intraneuronal inclusions consisting of hyperphosphorylated tau protein aggregates. Although Alzheimer's disease (AD) is also characterised by tau lesions, these are both histologically and biochemically distinct from the tau aggregates found in PiD. What determines the distinct characteristics of these tau lesions is unknown. As phosphorylated, soluble tau has been suggested to be the precursor of tau aggregates, we compared both the level and phosphorylation profile of tau in tissue extracts of AD and PiD brains to determine whether the differences in the tau lesions are reflected by differences in soluble tau. Levels of soluble tau were decreased in AD but not PiD. In addition, soluble tau was phosphorylated to a greater extent in AD than in PiD and displayed a different phosphorylation profile in the two disorders. Consistently, tau kinases were activated to different degrees in AD compared with PiD. Such differences in solubility and phosphorylation may contribute, at least in part, to the formation of distinct tau deposits, but may also have implications for the clinical differences between AD and PiD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Fosforilación , Enfermedad de Pick/metabolismo , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Western Blotting , Encéfalo/enzimología , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Enfermedad de Pick/enzimología , Lóbulo Temporal/metabolismo
12.
Nat Commun ; 8(1): 473, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28883427

RESUMEN

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


Asunto(s)
Lesiones Encefálicas/genética , Modelos Animales de Enfermedad , Accidente Cerebrovascular/genética , Proteínas tau/genética , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Transducción de Señal/genética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Sinaptosomas/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas tau/deficiencia
13.
PLoS One ; 11(10): e0163236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27736877

RESUMEN

Several mouse lines with knockout of the tau-encoding MAPT gene have been reported in the past; they received recent attention due to reports that tau reduction prevented Aß-induced deficits in mouse models of Alzheimer's disease. However, the effects of long-term depletion of tau in vivo remained controversial. Here, we used the tau-deficient GFP knockin line Mapttm1(EGFP)kit on a pure C57Bl/6 background and subjected a large cohort of males and females to a range of motor, memory and behavior tests and imaging analysis, at the advanced age of over 16 months. Neither heterozygous nor homozygous Mapttm1(EGFP)kit mice presented with deficits or abnormalities compared to wild-type littermates. Differences to reports using other tau knockout models may be due to different genetic backgrounds, respective gene targeting strategies or other confounding factors, such as nutrition. To this end, we report no functional or morphological deficits upon tau reduction or depletion in aged mice.


Asunto(s)
Enfermedad de Alzheimer/genética , Técnicas de Inactivación de Genes , Proteínas tau/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Locomoción , Masculino , Aprendizaje por Laberinto , Memoria , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
Neurosci Lett ; 631: 24-29, 2016 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-27521751

RESUMEN

Frontotemporal dementia (FTD) presents clinically with behavioral changes including disinhibition. Mutations in the tau-encoding MAPT gene identified in familial cases of FTD have been used to generate transgenic mouse models of the human condition. Here, we report behavioral changes in a recently developed P301S mutant tau transgenic mouse, including disinhibition-like behavior in the elevated plus maze and hyperactivity in the open field arena. Furthermore, histological analysis revealed the amygdala as a primary and early site of pathological tau deposition in these mice. Taken together, neuropathological and behavioral changes in P301S tau transgenic mice resemble features of human FTD.


Asunto(s)
Conducta Animal/fisiología , Demencia Frontotemporal/genética , Demencia Frontotemporal/psicología , Proteínas tau/genética , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Modelos Animales de Enfermedad , Humanos , Hipercinesia/genética , Masculino , Ratones , Ratones Transgénicos , Actividad Motora , Mutación , Proteínas tau/metabolismo
15.
Science ; 354(6314): 904-908, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27856911

RESUMEN

Amyloid-ß (Aß) toxicity in Alzheimer's disease (AD) is considered to be mediated by phosphorylated tau protein. In contrast, we found that, at least in early disease, site-specific phosphorylation of tau inhibited Aß toxicity. This specific tau phosphorylation was mediated by the neuronal p38 mitogen-activated protein kinase p38γ and interfered with postsynaptic excitotoxic signaling complexes engaged by Aß. Accordingly, depletion of p38γ exacerbated neuronal circuit aberrations, cognitive deficits, and premature lethality in a mouse model of AD, whereas increasing the activity of p38γ abolished these deficits. Furthermore, mimicking site-specific tau phosphorylation alleviated Aß-induced neuronal death and offered protection from excitotoxicity. Our work provides insights into postsynaptic processes in AD pathogenesis and challenges a purely pathogenic role of tau phosphorylation in neuronal toxicity.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Neurotoxinas/antagonistas & inhibidores , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Transducción de Señal
16.
PLoS One ; 6(12): e26860, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174735

RESUMEN

In Alzheimer's disease (AD) brains, the microtubule-associated protein tau and amyloid-ß (Aß) deposit as intracellular neurofibrillary tangles (NFTs) and extracellular plaques, respectively. Tau deposits are furthermore found in a significant number of frontotemporal dementia cases. These diseases are characterized by progressive neurodegeneration, the loss of intellectual capabilities and behavioral changes. Unfortunately, the currently available therapies are limited to symptomatic relief. While active immunization against Aß has shown efficacy in both various AD mouse models and patients with AD, immunization against pathogenic tau has only recently been shown to prevent pathology in young tau transgenic mice. However, if translated to humans, diagnosis and treatment would be routinely done when symptoms are overt, meaning that the histopathological changes have already progressed. Therefore, we used active immunization to target pathogenic tau in 4, 8, and 18 months-old P301L tau transgenic pR5 mice that have an onset of NFT pathology at 6 months of age. In all age groups, NFT pathology was significantly reduced in treated compared to control pR5 mice. Similarly, phosphorylation of tau at pathological sites was reduced. In addition, increased astrocytosis was found in the oldest treated group. Taken together, our data suggests that tau-targeted immunization slows the progression of NFT pathology in mice, with practical implications for human patients.


Asunto(s)
Envejecimiento/patología , Progresión de la Enfermedad , Inmunización , Ovillos Neurofibrilares/patología , Proteínas tau/inmunología , Secuencia de Aminoácidos , Animales , Gliosis/complicaciones , Gliosis/patología , Humanos , Inmunidad/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Fosforilación , Vacunación , Proteínas tau/química , Proteínas tau/metabolismo
17.
PLoS One ; 6(7): e22850, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829535

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are characterized by intraneuronal deposition of the nuclear TAR DNA-binding protein 43 (TDP-43) caused by unknown mechanisms. Here, we studied TDP-43 in primary neurons under different stress conditions and found that only proteasome inhibition by MG-132 or lactacystin could induce significant cytoplasmic accumulation of TDP-43, a histopathological hallmark in disease. This cytoplasmic accumulation was accompanied by phosphorylation, ubiquitination and aggregation of TDP-43, recapitulating major features of disease. Proteasome inhibition produced similar effects in both hippocampal and cortical neurons, as well as in immortalized motor neurons. To determine the contribution of TDP-43 to cell death, we reduced TDP-43 expression using small interfering RNA (siRNA), and found that reduced levels of TDP-43 dose-dependently rendered neurons more vulnerable to MG-132. Taken together, our data suggests a role for the proteasome in subcellular localization of TDP-43, and possibly in disease.


Asunto(s)
Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteasoma , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Leupeptinas/farmacología , Ratones , Neuronas/citología , Fosforilación , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Fracciones Subcelulares , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA