Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nature ; 480(7376): E4-5; discussion E6, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22158250

RESUMEN

Arising from C. J. Phiel, C. A. Wilson, V. M.-Y. Lee & P. S. Klein 423, 435-439 (2003)A major unresolved issue in Alzheimer's disease is identifying the mechanisms that regulate proteolytic processing of amyloid precursor protein (APP)-glycogen synthase kinase-3 (GSK-3) isozymes are thought to be important in this regulation. Phiel et al. proposed that GSK-3α, but not GSK-3ß, controls production of amyloid. We analysed the proteolytic processing of mouse and human APP in mouse brain in vivo in five different genetic and viral models. Our data do not yield evidence for either GSK-3α-mediated or GSK-3ß-mediated control of APP processing in brain in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Animales
2.
Neurobiol Dis ; 67: 119-32, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24704314

RESUMEN

Tau.P301L transgenic mice suffer precocious mortality between ages 8 and 11 months, resulting from upper airway defects caused by tauopathy in autonomic brainstem circuits that control breathing (Dutschmann et al., 2010). In individual mice, the clinical phenotype evolves progressively and rapidly (3-6weeks) from clasping, over general motor impairment to severe reduction in body-weight into the terminal phase that announces imminent death (<3days). Surprisingly, co-expression of GSK3ß with Tau.P301L significantly prolonged survival of bigenic biGT mice (Terwel et al., 2008), which we here assign to delayed development of brainstem tauopathy. Eventually, brainstem tauopathy became as prominent in old biGT mice in the specified brainstem nuclei as in the parental Tau.P301L mice, resulting in similar clinical deterioration and terminal phase preceding death, although at later age. Biochemically, in both genotypes the pathway to neurofibrillary tangles and neuropil threads was similar: phosphorylation of protein Tau and formation of soluble oligomers and insoluble aggregates, ending in the typical tangles and threads of tauopathy. The extra GSK3ß activity led to expected increased phosphorylation of protein Tau, particularly at residues S262 and S396, which we must conclude to delay the aggregation of protein Tau in the brainstem of aging biGT mice. The unexpected, paradoxical alleviation of the brainstem problems in biGT mice allowed them to grow older and thereby develop more severe tauopathy in forebrain than Tau.P301L mice, which succumb at younger age.


Asunto(s)
Tronco Encefálico/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Tauopatías/enzimología , Proteínas tau/química , Proteínas tau/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Tronco Encefálico/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Análisis de Supervivencia , Tauopatías/metabolismo
3.
Eur J Neurosci ; 40(2): 2442-53, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24754737

RESUMEN

The microtubule-associated protein Tau is responsible for a large group of neurodegenerative disorders, known as tauopathies, including Alzheimer's disease. Tauopathy result from augmented and/or aberrant phosphorylation of Tau. Besides aging and various genetic and epigenetic defects that remain largely unknown, an important non-genetic agent that contributes is hypothermia, eventually caused by anesthesia. Remarkably, tauopathy in brains of hibernating mammals is not pathogenic, and, because it is fully reversible, is even considered to be neuroprotective. Here, we assessed the terminal phase of Tau.P301L mice and bigenic crosses with mice lacking glycogen synthase kinase 3 (GSK3)α completely, or GSK3ß specifically in neurons. We also analysed biGT bigenic mice that co-express Tau.P301L with GSK3ß.S9A and develop severe forebrain tauopathy with age. We found that the precocious mortality of Tau.P301L mice was typified by hypothermia that aggravated Tau phosphorylation, but, surprisingly, independently of GSK3α/ß. The important contribution of hypothermia at the time of death of mice with tauopathy suggests that body temperature should be included as a parameter in the analysis of pre-clinical models, and, by extension, in patients suffering from tauopathy.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Procesamiento Proteico-Postraduccional , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Glucógeno Sintasa Quinasa 3/genética , Humanos , Hipotermia/metabolismo , Hipotermia/fisiopatología , Ratones , Neuronas/metabolismo , Fosforilación , Prosencéfalo/citología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Tauopatías/fisiopatología , Proteínas tau/genética , Proteínas tau/toxicidad
4.
Synapse ; 67(6): 313-27, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23390016

RESUMEN

Glycogen synthase kinase-3ß (GSK3ß) activity has been previously linked to Alzheimer's disease (AD) by its phosphorylation of tau and activation by amyloid. GSK3ß intracellular distribution is important in regulating its activity by restricting access to compartment-specific substrates. This study investigated regional and intracellular distribution of GSK3ß in a mouse model of AD, a bigenic mouse with combined amyloid and tau pathology (BiAT), and controls (FVB). At two different ages, the entire rostrocaudal extent of each brain was examined. Young (6-months-old) FVB and BiAT mice did not differ in GSK3ß expression and localization. In old (13-month-old) BiAT mice, neurons showed increased GSK3ß expression only in AD-relevant brain regions as compared with modest staining in region- and age-matched controls. Two regions with the most robust changes between FVB and BiAT mice, the amygdala and piriform cortex, were quantified at the light microscopic level. In both regions, the density of darkly labeled neurons was significantly greater in the old BiAT mice vs. the old FVB mice. Electron microscopy of the piriform cortex showed neuronal GSK3ß labeling in the rough endoplasmic reticulum, on ribosomes, and on microtubules in dendrites in both strains of mice. In old BiAT mice, GSK3ß labeling was qualitatively more robust compared to age-matched controls, and GSK3ß also appeared in neurofibrillary tangles. In conclusion, GSK3ß expression was increased in specific intracellular locations and was found in tangles in old BiAT mice, suggesting that GSK3ß overexpression in specific brain areas may be intrinsic to AD pathology.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Dendritas/metabolismo , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Expresión Génica , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Transgénicos , Microtúbulos/metabolismo , Ovillos Neurofibrilares/metabolismo , Ribosomas/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
5.
J Neurosci ; 31(49): 18036-47, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22159117

RESUMEN

Cognitive demise correlates with progressive brain tauopathy in dementing patients. Improved cognition of young Tau.P301L mice contrasts with dysfunction later in life and remains unexplained (Boekhoorn et al., 2006). To unravel early mechanisms, we composed a correlative time line of clinical symptoms, cognitive defects, and biochemical and pathological traits, including comprehensive analysis of dendritic spines in specified regions of the cortex and hippocampus of young and adult Tau.P301L mice. Remarkably, young Tau.P301L mice have not more, but more mature spines than wild-type mice, revealing the anatomical substrate for their improved cognition and LTP. Spine maturation remained high in the hippocampus of adult Tau.P301L mice. However, spines regressed in length paralleling impaired cognition and increased Tau phosphorylation (Terwel et al., 2005). Conversely, spine maturation was unaffected in adult Tau.4R mice, while spine density was increased and length reduced similar to Tau.P301L mice. To explain how protein Tau promoted spinogenesis, we analyzed hippocampal synaptosomes and dendritic spines for mouse and human Tau. While synaptosomes were positive for both mouse and human Tau, weak variable reaction in spines was observed only after fixation according to Bouin. Mouse Tau was absent from spines in wild-type mice, dissociating the pathological actions of Tau in transgenic mice by relocalization into dendrites and spines from the physiological actions of protein Tau in axons only. We conclude that mutant protein Tau modulates cognition and morphology of spines similarly and in both directions, with pathology later in life coinciding with increased phosphorylation and relocalization of Tau from axons to soma and processes.


Asunto(s)
Encéfalo/patología , Trastornos del Conocimiento , Espinas Dendríticas/patología , Leucina/genética , Neuronas/patología , Prolina/genética , Proteínas tau/genética , Factores de Edad , Animales , Proteínas Bacterianas/genética , Encéfalo/citología , Células Cultivadas , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Fosforilación/genética , Sinaptosomas/metabolismo , Proteínas tau/metabolismo
6.
Neurobiol Dis ; 46(1): 234-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22316605

RESUMEN

The postoperative cognitive decline resulting from volatile anesthesia is gaining acceptance as a major health problem. The common anesthetic isoflurane is suspected to precipitate neurodegeneration in Alzheimer's disease by unknown mechanisms. We previously validated that 8month old Tau.P301L mice suffer upper airways defects related to tauopathy within the Kolliker-Fuse nucleus that controls upper airways function. We now report that isoflurane anesthesia in young, pre-symptomatic Tau.P301L mice triggered precocious upper airways defects and tauopathy in several brainstem nuclei, including the nucleus ambiguus that contains upper airways motor neurons and the Kolliker-Fuse. The prescription drug memantine, identified as an NMDA receptor antagonist, prevented the post-anesthesia upper airways dysfunction and alleviated tauopathy in the nucleus ambiguus and Kolliker-Fuse. We further identified protocols of anesthesia in young Tau.P301L mice that mitigated adverse effects of isoflurane anesthesia. Thus, our experimental findings in a validated mouse model for tauopathy demonstrate the link between isoflurane anesthesia, earlier onset of tauopathy and earlier onset of functional deficits, highlight the implication of NMDA-receptors in the mechanisms mediating the adverse effects of isoflurane, and potentially identify safer protocols for anesthesia in patients with tauopathy.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Isoflurano/toxicidad , Degeneración Nerviosa/inducido químicamente , Insuficiencia Respiratoria/inducido químicamente , Tauopatías/inducido químicamente , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Animales , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/patología , Modelos Animales de Enfermedad , Ratones , Ratones Mutantes Neurológicos , Ratones Transgénicos , Degeneración Nerviosa/patología , Degeneración Nerviosa/prevención & control , Insuficiencia Respiratoria/patología , Insuficiencia Respiratoria/prevención & control , Tauopatías/patología , Tauopatías/prevención & control
7.
Am J Pathol ; 179(4): 2001-15, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21839061

RESUMEN

Adeno-associated virus (AAV)-mediated expression of wild-type or mutant P301L protein tau produces massive degeneration of pyramidal neurons without protein tau aggregation. We probed this novel model for genetic and structural factors and early parameters of pyramidal neurodegeneration. In yellow fluorescent protein-expressing transgenic mice, intracerebral injection of AAV-tauP301L revealed early damage to apical dendrites of CA1 pyramidal neurons, whereas their somata remained normal. Ultrastructurally, more and enlarged autophagic vacuoles were contained in degenerating dendrites and manifested as dark, discontinuous, vacuolated processes surrounded by activated astrocytes. Dendritic spines were lost in AAV-tauP301L-injected yellow fluorescent protein-expressing transgenic mice, and ultrastructurally, spines appeared dark and degenerating. In CX3CR1(EGFP/EGFP)-deficient mice, microglia were recruited early to neurons expressing human tau. The inflammatory response was accompanied by extravasation of plasma immunoglobulins. α2-Macroglobulin, but neither albumin nor transferrin, became lodged in the brain parenchyma. Large proteins, but not Evans blue, entered the brain of mice injected with AAV-tauP301L. Ultrastructurally, brain capillaries were constricted and surrounded by swollen astrocytes with extensions that contacted degenerating dendrites and axons. Together, these data corroborate the hypothesis that neuroinflammation participates essentially in tau-mediated neurodegeneration, and the model recapitulates early dendritic defects reminiscent of "dendritic amputation" in Alzheimer's disease.


Asunto(s)
Dendritas/patología , Inflamación/patología , Degeneración Nerviosa/patología , Sistema Nervioso/irrigación sanguínea , Sistema Nervioso/patología , Proteínas tau/metabolismo , Animales , Axones/patología , Axones/ultraestructura , Biomarcadores/metabolismo , Vasos Sanguíneos/patología , Vasos Sanguíneos/ultraestructura , Barrera Hematoencefálica/patología , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/ultraestructura , Dendritas/ultraestructura , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/complicaciones , Masculino , Ratones , Degeneración Nerviosa/complicaciones , Estrés Oxidativo , Permeabilidad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Sinapsis/patología , Sinapsis/ultraestructura
8.
J Neurosci ; 30(5): 1810-21, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20130190

RESUMEN

Tauopathy comprises hyperphosphorylation of the microtubule-associated protein tau, causing intracellular aggregation and accumulation as neurofibrillary tangles and neuropil treads. Some primary tauopathies are linked to mutations in the MAPT gene coding for protein tau, but most are sporadic with unknown causes. Also, in Alzheimer's disease, the most frequent secondary tauopathy, neither the cause nor the pathological mechanisms and repercussions are understood. Transgenic mice expressing mutant Tau-P301L suffer cognitive and motor defects and die prematurely from unknown causes. Here, in situ electrophysiology in symptomatic Tau-P301L mice (7-8 months of age) revealed reduced postinspiratory discharges of laryngeal motor outputs that control laryngeal constrictor muscles. Under high chemical drive (hypercapnia), postinspiratory discharge was nearly abolished, whereas laryngeal inspiratory discharge was increased disproportionally. The latter may suggest a shift of postinspiratory laryngeal constrictor activity into inspiration. In vivo double-chamber plethysmography of Tau-P301L mice showed significantly reduced respiratory airflow but significantly increased chest movements during baseline breathing, but particularly in hypercapnia, confirming a significant increase in inspiratory resistive load. Histological analysis demonstrated hyperphosphorylated tau in brainstem nuclei, directly or indirectly involved in upper airway motor control (i.e., the Kölliker-Fuse, periaqueductal gray, and intermediate reticular nuclei). In contrast, young Tau-P301L mice did not show breathing disorders or brainstem tauopathy. Consequently, in aging Tau-P301L mice, progressive upper airway dysfunction is linked to progressive tauopathy in identified neural circuits. Because patients with tauopathy suffer from upper airway dysfunction, the Tau-P301L mice can serve as an experimental model to study disease-specific synaptic dysfunction in well defined functional neural circuits.


Asunto(s)
Tronco Encefálico/metabolismo , Mesencéfalo/metabolismo , Trastornos Respiratorios/genética , Trastornos Respiratorios/patología , Tauopatías/complicaciones , Tauopatías/patología , Proteínas tau/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Tronco Encefálico/patología , Modelos Animales de Enfermedad , Mesencéfalo/patología , Ratones , Ratones Transgénicos , Mutación , Fosforilación , Pletismografía , Ventilación Pulmonar , Trastornos Respiratorios/fisiopatología , Proteínas tau/metabolismo
9.
Biochim Biophys Acta ; 1802(10): 808-18, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20332023

RESUMEN

Alzheimer's dementia is developing ever more as a complex syndrome with various unknown genetic and epigenetic contributions. These are compounded on and exacerbating the underlying amyloid and tau pathology that remain the basis of the pathological definition of Alzheimer's disease. Here, we present a selection of aspects of recent bigenic and virus-based mouse strains, developed as pre-clinical models for Alzheimer's disease. We discuss newer features in the context of the characteristics defined in previously validated transgenic models. We focus on specific aspects of single and multiple transgenic mouse models for Alzheimer's disease and for tauopathies, rather than providing an exhaustive list of all available models. We concentrate on the content of information related to neurodegeneration and disease mechanisms. We pay attention to aspects and defects that are predicted by the models and can be tested in humans. We discuss implications that help translate the fundamental knowledge into clinical, diagnostic and therapeutic applications. We elaborate on the increasing knowledge extracted from transgenic models and from newer adeno-associated viral models. We advocate this combination as a valuable strategy to study molecular, cellular and system-related pathogenic mechanisms in AD and tauopathies. We believe that innovative animal models remain needed to critically test current views, to identify and validate therapeutic targets, to allow testing of compounds, to help understand and eventually treat tauopathies, including Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/virología , Modelos Animales de Enfermedad , Tauopatías/genética , Tauopatías/virología , Enfermedad de Alzheimer/patología , Animales , Humanos , Ratones , Ratones Transgénicos , Tauopatías/patología
10.
Neurobiol Dis ; 44(3): 284-91, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21807097

RESUMEN

APP.V717I and Tau.P301L transgenic mice develop Alzheimer's disease pathology comprising important aspects of human disease including increased levels of amyloid peptides, cognitive and motor impairment, amyloid plaques and neurofibrillary tangles. The combined model, APP.V717I×Tau.P301L bigenic mice (biAT mice) exhibit aggravated amyloid and tau pathology with severe cognitive and behavioral defects. In the present study, we investigated early changes in synaptic function in the CA1 and CA3 regions of acute hippocampal slices of young APP.V717I, Tau.P301L and biAT transgenic animals. We have used planar multi-electrode arrays (MEA) and improved methods for simultaneous multi-site recordings from two hippocampal sub-regions. In the CA1 region, long-term potentiation (LTP) was severely impaired in all transgenic animals when compared with age-matched wild-type controls, while basal synaptic transmission and paired-pulse facilitation were minimally affected. In the CA3 region, LTP was normal in Tau.P301L and APP.V717I but clearly impaired in biAT mice. Surprisingly, frequency facilitation in CA3 was significantly enhanced in Tau.P301L mice, while not affected in APP.V717I mice and depressed in biAT mice. The findings demonstrate important synaptic changes that differ considerably in the hippocampal sub-regions already at young age, well before the typical amyloid or tau pathology is evident.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Electrodos , Hipocampo/patología , Hipocampo/fisiopatología , Sinapsis/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Biofisica , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Humanos , Técnicas In Vitro , Isoleucina/genética , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Mutación/genética , Sinapsis/fisiología , Factores de Tiempo , Valina/genética , Proteínas tau/genética
11.
EBioMedicine ; 39: 377-387, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30502054

RESUMEN

BACKGROUND: Glycogen synthase kinase-3ß (GSK3ß) is a key regulator of cellular homeostasis. In neurons, GSK3ß contributes to the control of neuronal transmission and plasticity, but its role in epilepsy remains to be defined. METHODS: Biochemical and electrophysiological methods were used to assess the role of GSK3ß in regulating neuronal transmission and epileptogenesis. GSK3ß activity was increased genetically in GSK3ß[S9A] mice. Its effects on neuronal transmission and epileptogenesis induced by kainic acid were assessed by field potential recordings in mice brain slices and video electroencephalography in vivo. The ion channel expression was measured in brain samples from mice and followed by analysis in samples from patients with temporal lobe epilepsy or focal cortical dysplasia in correlation to GSK3ß phosphorylation. FINDINGS: Higher GSK3ß activity decreased the progression of kainic acid induced epileptogenesis. At the biochemical level, higher GSK3ß activity increased the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel 4 under basal conditions and in the epileptic mouse brain and decreased phosphorylation of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 at Serine 831 under basal conditions. Moreover, we found a significant correlation between higher inhibitory GSK3ß phosphorylation at Serine 9 and higher activating GluA1 phosphorylation at Serine 845 in brain samples from epileptic patients. INTERPRETATION: Our data imply GSK3ß activity in the protection of neuronal networks from hyper-activation in response to epileptogenic stimuli and indicate that the anti-epileptogenic function of GSK3ß involves modulation of HCN4 level and the synaptic AMPA receptors pool.


Asunto(s)
Epilepsia/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Ácido Kaínico/efectos adversos , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Receptores AMPA/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Preescolar , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/inducido químicamente , Epilepsia/genética , Femenino , Glucógeno Sintasa Quinasa 3 beta/química , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fosforilación , Receptores AMPA/química , Transducción de Señal , Transmisión Sináptica , Grabación en Video
12.
Neurosci Lett ; 435(3): 186-9, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18343582

RESUMEN

Beta-site amyloid precursor protein-cleaving enzyme-1 (BACE1) initiates generation of amyloid beta (Abeta), a pathological hallmark of Alzheimer's disease. We investigated the impact of BACE1 protein level on endogenous Abeta. Endogenous Abeta and BACE1 protein levels were concurrently and significantly reduced during early life. However, Abeta levels were similar between BACE1 transgenic and wildtype mice. This suggests that BACE1 protein level has a minimal effect on the level of endogenous Abeta. Consequently, other factors must be involved in modulation of Abeta production in adult and ageing brain and investigation of such factors may yield therapeutic targets. Further, these results suggest that substantial inhibition of BACE1 in brain may be required for clinical benefit in Alzheimer's disease.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Factores de Edad , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Mol Neurobiol ; 55(7): 6050-6062, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29143288

RESUMEN

Glycogen synthase kinases-3ß (GSK3ß) is a key regulator of cell homeostasis. In neurons, GSK3ß contributes to control of neuronal transmission and plasticity. Despite extensive studies in non-neuronal cells, crosstalk between GSK3ß and other signaling pathways remains not well defined in neurons. In the present study, we report that GSK3ß positively affected the activity of effectors of mammalian target of rapamycin complex 1 (mTORC1) and complex 2 (mTORC2), in mature neurons in vitro and in vivo. GSK3ß also promoted prosurvival signaling and attenuated kainic acid-induced apoptosis. Our study identified GSK3ß as a positive regulator of prosurvival signaling, including the mTOR pathway, and indicates the possible neuroprotective role of GSK3ß in models of pharmacologically induced excitotoxicity.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neuronas/citología , Neuronas/enzimología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis , Encéfalo/enzimología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Isoenzimas/metabolismo , Ácido Kaínico , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína S6 Ribosómica/metabolismo
14.
Mol Neurobiol ; 54(1): 200-211, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26738851

RESUMEN

Changes in the morphology of dendritic spines are prominent during learning and in different neurological and neuropsychiatric diseases, including those in which glycogen synthase kinase-3ß (GSK-3ß) has been implicated. Despite much evidence of the involvement of GSK-3ß in functional synaptic plasticity, it is unclear how GSK-3ß controls structural synaptic plasticity (i.e., the number and shape of dendritic spines). In the present study, we used two mouse models overexpressing and lacking GSK-3ß in neurons to investigate how GSK-3ß affects the structural plasticity of dendritic spines. Following visualization of dendritic spines with DiI dye, we found that increasing GSK-3ß activity increased the number of thin spines, whereas lacking GSK-3ß increased the number of stubby spines in the dentate gyrus. Under conditions of neuronal excitation, increasing GSK-3ß activity caused higher activity of extracellularly acting matrix metalloproteinase-9 (MMP-9), and MMP inhibition normalized thin spines in GSK-3ß overexpressing mice. Administration of the nonspecific GSK-3ß inhibitor lithium in animals with active MMP-9 and animals lacking MMP-9 revealed that GSK-3ß and MMP-9 act in concert to control dendritic spine morphology. Altogether, our data demonstrate that the dysregulation of GSK-3ß activity has dramatic consequences on dendritic spine morphology, implicating MMP-9 as a mediator of GSK-3ß-induced synaptic alterations.


Asunto(s)
Espinas Dendríticas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Transgénicas , Ratas Wistar
15.
J Alzheimers Dis ; 56(2): 585-599, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28035925

RESUMEN

The microtubule-associated protein Tau is an intrinsically unfolded, very soluble neuronal protein. Under still unknown circumstances, Tau protein forms soluble oligomers and insoluble aggregates that are closely linked to the cause and progression of various brain pathologies, including Alzheimer's disease. Previously we reported the development of liposome-based vaccines and their efficacy and safety in preclinical mouse models for tauopathy. Here we report the use of a liposomal vaccine for the generation of a monoclonal antibody with particular characteristics that makes it a valuable tool for fundamental studies as well as a candidate antibody for diagnostic and therapeutic applications. The specificity and affinity of antibody ACI-5400 were characterized by a panel of methods: (i) measuring the selectivity for a specific phospho-Tau epitope known to be associated with tauopathy, (ii) performing a combination of peptide and protein binding assays, (iii) staining of brain sections from mouse preclinical tauopathy models and from human subjects representing six different tauopathies, and (iv) evaluating the selective binding to pathological epitopes on extracts from tauopathy brains in non-denaturing sandwich assays. We conclude that the ACI-5400 antibody binds to protein Tau phosphorylated at S396 and favors a conformation that is typically present in the brain of tauopathy patients, including Alzheimer's disease.


Asunto(s)
Anticuerpos Monoclonales , Tauopatías/diagnóstico , Tauopatías/terapia , Proteínas tau/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Epítopos , Humanos , Hibridomas , Liposomas , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Hilos del Neurópilo/metabolismo , Hilos del Neurópilo/patología , Fosforilación , Unión Proteica , Proteínas Recombinantes/inmunología , Tauopatías/inmunología , Tauopatías/patología , Vacunas
16.
PLoS One ; 9(2): e87605, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498342

RESUMEN

The stratum lacunosum moleculare (SLM) is the connection hub between entorhinal cortex and hippocampus, two brain regions that are most vulnerable in Alzheimer's disease. We recently identified a specific synaptic deficit of Nectin-3 in transgenic models for tauopathy. Here we defined cognitive impairment and electrophysiological problems in the SLM of Tau.P301L mice, which corroborated the structural defects in synapses and dendritic spines. Reduced diffusion of DiI from the ERC to the hippocampus indicated defective myelinated axonal pathways. Ultrastructurally, myelinated axons in the temporoammonic pathway (TA) that connects ERC to CA1 were damaged in Tau.P301L mice at young age. Unexpectedly, the myelin defects were even more severe in bigenic biGT mice that co-express GSK3ß with Tau.P301L in neurons. Combined, our data demonstrate that neuronal expression of protein Tau profoundly affected the functional and structural organization of the entorhinal-hippocampal complex, in particular synapses and myelinated axons in the SLM. White matter pathology deserves further attention in patients suffering from tauopathy and Alzheimer's disease.


Asunto(s)
Axones/metabolismo , Encéfalo/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Sinapsis/metabolismo , Tauopatías/genética , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Axones/patología , Axones/ultraestructura , Encéfalo/patología , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Corteza Entorrinal/metabolismo , Corteza Entorrinal/patología , Corteza Entorrinal/fisiopatología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Microscopía Electrónica , Actividad Motora/fisiología , Mutación , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/ultraestructura , Sinapsis/patología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Tauopatías/metabolismo , Tauopatías/fisiopatología , Proteínas tau/metabolismo
17.
Behav Brain Res ; 248: 46-50, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23570858

RESUMEN

Glycogen synthase kinase-3 (GSK-3), a multifunctional serine-threonine kinase, is an important regulator in numerous signaling pathways and processes including adult brain neurogenesis. GSK-3 (mal)functioning was implicated in many diseases, in particular neurological and behavioral disorders. We investigated the impact of altered levels of the GSK-3ß isoform on hippocampal size, number of doublecortin-positive cells, and hippocampal-dependent behaviors. Both GSK-3ß transgenic mice (GSK-3ß[S9A] mice) and GSK-3ß neuron-specific knockout (GSK-3ß(n-/-)) mice, showed reduced size of the dentate gyrus (DG) and were impaired in three hippocampal-dependent, species-typical behavioral tasks: digging, marble burying and nest building. We further demonstrate that the number of differentiating, doublecortin-positive new neurons is reduced in GSK-3ß[S9A] mice, but not in GSK-3ß(n-/-) mice. We conclude that GSK-3ß activity must be critically controlled to allow wild type-like volume of the dentate gyrus and for normal execution of hippocampal-dependent, species-typical behavior.


Asunto(s)
Conducta Animal/fisiología , Giro Dentado/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Ratones Noqueados/metabolismo , Ratones Transgénicos/metabolismo , Animales , Glucógeno Sintasa Quinasa 3 beta , Ratones , Neurogénesis/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología
18.
PLoS One ; 8(5): e63589, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23704923

RESUMEN

Cell adhesion molecules are important structural substrates, required for synaptic plasticity and synaptogenesis. CAMs differ widely in their expression throughout different brain regions and their specific structural and functional roles in the brain remain to be elucidated. Here, we investigated selected cell adhesion molecules for alterations in expression levels and neuronal localization in validated mouse models for Alzheimer's disease that mimic the age-related progression of amyloid accumulation and tauopathy. Among the cell adhesion molecules analyzed, Nectin-3 expression was affected most and specifically in all mouse models with tauopathy. In particular was Nectin-3 depleted from the specific region of the hippocampus, known as the stratum lacunosum and moleculare, in mice that express wild-type or mutant human protein Tau, either chronically or sub-acutely. Tauopathy progresses from the entorhinal cortex to the hippocampus by unknown mechanisms that could involve transport by the myelinated axons of the temporoammonic and perforant pathways. The decreased expression of Nectin-3 in the stratum lacunosum moleculare is an early marker of impaired transport, and eventual synaptic problems, caused by beginning tauopathy.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Moléculas de Adhesión Celular/metabolismo , Regulación hacia Abajo/genética , Tauopatías/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Nectinas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sinaptosomas/metabolismo , Tauopatías/patología
19.
Mol Brain ; 6: 27, 2013 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-23705847

RESUMEN

BACKGROUND: GSK3ß is involved in a wide range of physiological functions, and is presumed to act in the pathogenesis of neurological diseases, from bipolar disorder to Alzheimer's disease (AD). In contrast, the GSK3α isozyme remained largely ignored with respect to both aspects. RESULTS: We generated and characterized two mouse strains with neuron-specific or with total GSK3α deficiency. Behavioral and electrophysiological analysis demonstrated the physiological importance of neuronal GSK3α, with GSK3ß not compensating for impaired cognition and reduced LTP. Interestingly, the passive inhibitory avoidance task proved to modulate the phosphorylation status of both GSK3 isozymes in wild-type mice, further implying both to function in cognition. Moreover, GSK3α contributed to the neuronal architecture of the hippocampal CA1 sub-region that is most vulnerable in AD. Consequently, practically all parameters and characteristics indicated that both GSK3 isoforms were regulated independently, but that they acted on the same physiological functions in learning and memory, in mobility and in behavior. CONCLUSIONS: GSK3α proved to be regulated independently from GSK3ß, and to exert non-redundant physiological neurological functions in general behavior and in cognition. Moreover, GSK3α contributes to the pathological phosphorylation of protein Tau.


Asunto(s)
Cognición/fisiología , Pleiotropía Genética , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas tau/metabolismo , Animales , Conducta Animal , Peso Corporal , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Región CA1 Hipocampal/enzimología , Región CA1 Hipocampal/patología , Cruzamientos Genéticos , Femenino , Genotipo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Infertilidad Masculina/enzimología , Infertilidad Masculina/patología , Integrasas/metabolismo , Isoenzimas/metabolismo , Estimación de Kaplan-Meier , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Proteínas Mutantes/metabolismo , Neuronas/enzimología , Neuronas/patología , Tamaño de los Órganos , Especificidad de Órganos , Fenotipo , Fosforilación
20.
PLoS One ; 8(12): e84442, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376810

RESUMEN

The microtubule associated protein tau causes primary and secondary tauopathies by unknown molecular mechanisms. Post-translational O-GlcNAc-ylation of brain proteins was demonstrated here to be beneficial for Tau.P301L mice by pharmacological inhibition of O-GlcNAc-ase. Chronic treatment of ageing Tau.P301L mice mitigated their loss in body-weight and improved their motor deficits, while the survival was 3-fold higher at the pre-fixed study endpoint at age 9.5 months. Moreover, O-GlcNAc-ase inhibition significantly improved the breathing parameters of Tau.P301L mice, which underpinned pharmacologically the close correlation of mortality and upper-airway defects. O-GlcNAc-ylation of brain proteins increased rapidly and stably by systemic inhibition of O-GlcNAc-ase. Conversely, biochemical evidence for protein Tau.P301L to become O-GlcNAc-ylated was not obtained, nor was its phosphorylation consistently or markedly affected. We conclude that increasing O-GlcNAc-ylation of brain proteins improved the clinical condition and prolonged the survival of ageing Tau.P301L mice, but not by direct biochemical action on protein tau. The pharmacological effect is proposed to be located downstream in the pathological cascade initiated by protein Tau.P301L, opening novel venues for our understanding, and eventually treating the neurodegeneration mediated by protein tau.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Piranos/farmacología , Mecánica Respiratoria/fisiología , Tauopatías/tratamiento farmacológico , Tauopatías/fisiopatología , Tiazoles/farmacología , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , Análisis de Varianza , Animales , Western Blotting , Femenino , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Transgénicos , Pletismografía , Piranos/síntesis química , Mecánica Respiratoria/efectos de los fármacos , Tiazoles/síntesis química , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA