Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Am Heart Assoc ; 8(9): e011630, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31057061

RESUMEN

Background Arterial stiffness is associated with cognitive decline and dementia; however, the precise mechanisms by which it affects the brain remain unclear. Methods and Results Using a mouse model based on carotid calcification this study characterized mechanisms that could contribute to brain degeneration due to arterial stiffness. At 2 weeks postcalcification, carotid stiffness attenuated resting cerebral blood flow in several brain regions including the perirhinal/entorhinal cortex, hippocampus, and thalamus, determined by autoradiography ( P<0.05). Carotid calcification impaired cerebral autoregulation and diminished cerebral blood flow responses to neuronal activity and to acetylcholine, examined by laser Doppler flowmetry ( P<0.05, P<0.01). Carotid stiffness significantly affected spatial memory at 3 weeks ( P<0.05), but not at 2 weeks, suggesting that cerebrovascular impairments precede cognitive dysfunction. In line with the endothelial deficits, carotid stiffness led to increased blood-brain barrier permeability in the hippocampus ( P<0.01). This region also exhibited reductions in vessel number containing collagen IV ( P<0.01), as did the somatosensory cortex ( P<0.05). No evidence of cerebral microhemorrhages was present. Carotid stiffness did not affect the production of mouse amyloid-ß (Aß) or tau phosphorylation, although it led to a modest increase in the Aß40/Aß42 ratio in frontal cortex ( P<0.01). Conclusions These findings suggest that carotid stiffness alters brain microcirculation and increases blood-brain barrier permeability associated with cognitive impairments. Therefore, arterial stiffness should be considered a relevant target to protect the brain and prevent cognitive dysfunctions.


Asunto(s)
Conducta Animal , Encéfalo/irrigación sanguínea , Arterias Carótidas/fisiopatología , Enfermedades de las Arterias Carótidas/complicaciones , Circulación Cerebrovascular , Cognición , Disfunción Cognitiva/etiología , Calcificación Vascular/complicaciones , Rigidez Vascular , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Enfermedades de las Arterias Carótidas/fisiopatología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Fragmentos de Péptidos/metabolismo , Memoria Espacial , Factores de Tiempo , Calcificación Vascular/fisiopatología , Proteínas tau/metabolismo
2.
Brain Res ; 1711: 106-114, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30641037

RESUMEN

Parkinson's disease (PD) is characterized by motor deficits, although cognitive disturbances are frequent and have been noted early in the disease. The main pathological characteristics of PD are the loss of dopaminergic neurons and the presence of aggregated α-synuclein in Lewy bodies of surviving cells. Studies have also documented the presence of other proteins within Lewy bodies, particularly tau, a microtubule-associated protein implicated in a wide range of neurodegenerative diseases, including Alzheimer's disease (AD). In AD, tau pathology correlates with cognitive dysfunction, and tau mutations have been reported to lead to dementia associated with parkinsonism. However, the role of tau in PD pathogenesis remains unclear. To address this question, we induced parkinsonism by injecting the toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in hTau mice, a mouse model of tauopathy expressing human tau, and a mouse model knock-out for tau (TKO). We found that although MPTP impaired locomotion (gait analysis) and cognition (Barnes maze), there were no discernable differences between hTau and TKO mice. MPTP also induced a slight but significant increase in tau phosphorylation (Thr205) in the hippocampus of hTau mice, as well as a significant decrease in the soluble and insoluble tau fractions that correlated with the loss of dopaminergic neurons in the brainstem. Overall, our findings suggest that, although MPTP can induce an increase in tau phosphorylation at specific epitopes, tau does not seem to causally contribute to cognitive and locomotor deficits induced by this toxin.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Cognición/fisiología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fosforilación , Tauopatías/fisiopatología , alfa-Sinucleína/metabolismo
3.
Sci Rep ; 7: 46359, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28402338

RESUMEN

Abnormally hyperphosphorylated tau aggregated as intraneuronal neurofibrillary tangles is one of the two neuropathological hallmarks of Alzheimer's disease (AD). The majority of AD cases are sporadic with numerous environmental, biological and genetic risks factors. Interestingly, insulin dysfunction and hyperglycaemia are both risk factors for sporadic AD. However, how hyperglycaemia and insulin dysfunction affect tau pathology, is not well understood. In this study, we examined the effects of insulin deficiency on tau pathology in transgenic hTau mice by injecting different doses of streptozotocin (STZ), a toxin that destroys insulin-producing cells in the pancreas. One high dose of STZ resulted in marked diabetes, and five low doses led to a milder diabetes. Both groups exhibited brain tau hyperphosphorylation but no increased aggregation. Tau hyperphosphorylation correlated with inhibition of Protein Phosphatase 2A (PP2A), the main tau phosphatase. Interestingly, insulin injection 30 minutes before sacrifice partially restored tau phosphorylation to control levels in both STZ-injected groups. Our results confirm a link between insulin homeostasis and tau phosphorylation, which could explain, at least in part, a higher incidence of AD in diabetic patients.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Proteína Fosfatasa 2/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Tauopatías/complicaciones , Tauopatías/patología
4.
Methods Mol Biol ; 1523: 263-272, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27975255

RESUMEN

In Alzheimer's disease and other tauopathies, tau displays several abnormal post-translation modifications such as hyperphosphorylation, truncation, conformation, and oligomerization. Mouse monoclonal antibodies have been raised against such tau modifications for research, diagnostic, and therapeutic purposes. However, many of these primary antibodies are at risk of giving nonspecific signals in common Western blotting procedures. Not because they are unspecific, but because the secondary antibodies used to detect them will also detect the heavy chain of endogenous mouse immunoglobulins (Igs), and give a nonspecific signal at the same molecular weight than tau protein (around 50 kDa). Here, we propose the use of anti-light chain secondary antibodies as a simple and efficient technique to prevent nonspecific Igs signals at around 50 kDa. We demonstrate the efficacy of this method by removing artifactual signals when using monoclonal antibodies directed at tau phosphorylation (AT100, 12E8, AT270), tau truncation (TauC3), tau oligomerization (TOMA), or tau abnormal conformation (Alz50), in wild-type, 3×Tg-AD, and tau knockout mice.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Encéfalo/metabolismo , Proteínas tau/metabolismo , Animales , Especificidad de Anticuerpos , Western Blotting , Cadenas Ligeras de Inmunoglobulina/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Mutación Puntual , Proteínas tau/genética
5.
Neurobiol Aging ; 36(8): 2414-28, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26058840

RESUMEN

There is developing interest in the potential association between anesthesia and the onset and progression of Alzheimer's disease. Several anesthetics have, thus, been demonstrated to induce tau hyperphosphorylation, an effect mostly mediated by anesthesia-induced hypothermia. Here, we tested the hypothesis that acute normothermic administration of dexmedetomidine (Dex), an intravenous sedative used in intensive care units, would result in tau hyperphosphorylation in vivo and in vitro. When administered to nontransgenic mice, Dex-induced tau hyperphosphorylation persisting up to 6 hours in the hippocampus for the AT8 epitope. Pretreatment with atipamezole, a highly specific α2-adrenergic receptor antagonist, blocked Dex-induced tau hyperphosphorylation. Furthermore, Dex dose-dependently increased tau phosphorylation at AT8 in SH-SY5Y cells, impaired mice spatial memory in the Barnes maze and promoted tau hyperphosphorylation and aggregation in transgenic hTau mice. These findings suggest that Dex: (1) increases tau phosphorylation, in vivo and in vitro, in the absence of anesthetic-induced hypothermia and through α2-adrenergic receptor activation, (2) promotes tau aggregation in a mouse model of tauopathy, and (3) impacts spatial reference memory.


Asunto(s)
Dexmedetomidina/efectos adversos , Hipnóticos y Sedantes/efectos adversos , Proteínas tau/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Células Cultivadas , Dexmedetomidina/administración & dosificación , Dexmedetomidina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Hipocampo/metabolismo , Humanos , Hipnóticos y Sedantes/administración & dosificación , Hipotermia Inducida , Técnicas In Vitro , Infusiones Intravenosas , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Agregación Patológica de Proteínas/inducido químicamente , Memoria Espacial/efectos de los fármacos
6.
Neurobiol Aging ; 36(2): 901-2, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25491074

RESUMEN

Alzheimer's disease is characterized by the deposition of intracellular aggregates of hyperphosphorylated tau protein. Tau hyperphosphorylation has been attributed in part to the deregulation of kinases and phosphatases activities. Extracellular signal regulated-kinases 1/2 (ERK1/2) were reported to be activated in the first stages of Alzheimer's disease and were proposed as a potential therapeutic target. However, although the phosphorylation of tau by ERK1/2 has been demonstrated in cell-free system, it remains controversial in vivo. Here, we showed that pharmacologic inhibition of ERK1/2 in mice and SH-SY5Y cells did not reduce basal levels of phospho-tau or hypothermia-induced tau hyperphosphorylation. We also found that activating ERK1/2 by hyperthermia did not correlate with increased tau phosphorylation. Finally, ERK1/2 was inhibited, but tau phosphorylation was not altered in Mek1-/- mice. In conclusion, these results do not support the involvement of ERK1/2 in tau phosphorylation under physiological conditions.


Asunto(s)
Enfermedad de Alzheimer/etiología , Sistema de Señalización de MAP Quinasas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/terapia , Animales , Células Cultivadas , Humanos , Hipertermia Inducida , Hipotermia Inducida , Técnicas In Vitro , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Fosforilación
7.
PLoS One ; 9(5): e94251, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24788298

RESUMEN

Aggregates of hyperphosphorylated tau protein are found in a group of diseases called tauopathies, which includes Alzheimer's disease. The causes and consequences of tau hyperphosphorylation are routinely investigated in laboratory animals. Mice are the models of choice as they are easily amenable to transgenic technology; consequently, their tau phosphorylation levels are frequently monitored by Western blotting using a panel of monoclonal/polyclonal anti-tau antibodies. Given that mouse secondary antibodies can recognize endogenous mouse immunoglobulins (Igs) and the possible lack of specificity with some polyclonal antibodies, non-specific signals are commonly observed. Here, we characterized the profiles of commonly used anti-tau antibodies in four different mouse models: non-transgenic mice, tau knock-out (TKO) mice, 3xTg-AD mice, and hypothermic mice, the latter a positive control for tau hyperphosphorylation. We identified 3 tau monoclonal antibody categories: type 1, characterized by high non-specificity (AT8, AT180, MC1, MC6, TG-3), type 2, demonstrating low non-specificity (AT270, CP13, CP27, Tau12, TG5), and type 3, with no non-specific signal (DA9, PHF-1, Tau1, Tau46). For polyclonal anti-tau antibodies, some displayed non-specificity (pS262, pS409) while others did not (pS199, pT205, pS396, pS404, pS422, A0024). With monoclonal antibodies, most of the interfering signal was due to endogenous Igs and could be eliminated by different techniques: i) using secondary antibodies designed to bind only non-denatured Igs, ii) preparation of a heat-stable fraction, iii) clearing Igs from the homogenates, and iv) using secondary antibodies that only bind the light chain of Igs. All of these techniques removed the non-specific signal; however, the first and the last methods were easier and more reliable. Overall, our study demonstrates a high risk of artefactual signal when performing Western blotting with routinely used anti-tau antibodies, and proposes several solutions to avoid non-specific results. We strongly recommend the use of negative (i.e., TKO) and positive (i.e., hypothermic) controls in all experiments.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Modelos Animales de Enfermedad , Proteínas tau/inmunología , Secuencia de Aminoácidos , Animales , Artefactos , Técnicas de Inactivación de Genes , Humanos , Cadenas Ligeras de Inmunoglobulina/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
8.
Sci Rep ; 3: 1388, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24045785

RESUMEN

Several anesthetics have been reported to suppress the transcription of a number of genes, including Arc, also known as Arg3.1, an immediate early gene that plays a significant role in memory consolidation. The purpose of this study was to explore the mechanism of anesthesia-mediated depression in Arc gene and protein expression. Here, we demonstrate that isoflurane or propofol anesthesia decreases hippocampal Arc protein expression in rats and mice. Surprisingly, this change was secondary to anesthesia-induced hypothermia. Furthermore, we confirm in vivo and in vitro that hypothermia per se is directly responsible for decreased Arc protein levels. This effect was the result of the decline of Arc mRNA basal levels following inhibition of ERK/MAPK by hypothermia. Overall, our results suggest that anesthesia-induced hypothermia leads to ERK inhibition, which in turns decreases Arc levels. These data give new mechanistic insights on the regulation of immediate early genes by anesthesia and hypothermia.


Asunto(s)
Anestesia , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Hipotermia Inducida , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Anestésicos por Inhalación/farmacología , Animales , Línea Celular , Quinasa del Factor 2 de Elongación/metabolismo , Activación Enzimática/efectos de los fármacos , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isoflurano/farmacología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratas , Transducción de Señal , Transcripción Genética
9.
Diabetes ; 62(2): 609-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22961084

RESUMEN

The histopathological hallmarks of Alzheimer disease (AD) include intraneuronal neurofibrillary tangles composed of abnormally hyperphosphorylated τ protein. Insulin dysfunction might influence AD pathology, as population-based and cohort studies have detected higher AD incidence rates in diabetic patients. But how diabetes affects τ pathology is not fully understood. In this study, we investigated the impact of insulin dysfunction on τ phosphorylation in a genetic model of spontaneous type 1 diabetes: the nonobese diabetic (NOD) mouse. Brains of young and adult female NOD mice were examined, but young NOD mice did not display τ hyperphosphorylation. τ phosphorylation at τ-1 and pS422 epitopes was slightly increased in nondiabetic adult NOD mice. At the onset of diabetes, τ was hyperphosphorylated at the τ-1, AT8, CP13, pS262, and pS422. A subpopulation of diabetic NOD mice became hypothermic, and τ hyperphosphorylation further extended to paired helical filament-1 and TG3 epitopes. Furthermore, elevated τ phosphorylation correlated with an inhibition of protein phosphatase 2A (PP2A) activity. Our data indicate that insulin dysfunction in NOD mice leads to AD-like τ hyperphosphorylation in the brain, with molecular mechanisms likely involving a deregulation of PP2A. This model may be a useful tool to address further mechanistic association between insulin dysfunction and AD pathology.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Química Encefálica , Modelos Animales de Enfermedad , Femenino , Hipotermia , Ratones , Ratones Endogámicos NOD , Ovillos Neurofibrilares/metabolismo , Fosforilación , Isoformas de Proteínas/metabolismo
10.
Sci Rep ; 2: 480, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761989

RESUMEN

Tau hyperphosphorylation is one hallmark of Alzheimer's disease (AD) pathology. Pharmaceutical companies have thus developed kinase inhibitors aiming to reduce tau hyperphosphorylation. One obstacle in screening for tau kinase inhibitors is the low phosphorylation levels of AD-related phospho-epitopes in normal adult mice and cultured cells. We have shown that hypothermia induces tau hyperphosphorylation in vitro and in vivo. Here, we hypothesized that hypothermia could be used to assess tau kinase inhibitors efficacy. Hypothermia applied to models of biological gradual complexity such as neuronal-like cells, ex vivo brain slices and adult non-transgenic mice leads to tau hyperphosphorylation at multiple AD-related phospho-epitopes. We show that Glycogen Synthase Kinase-3 inhibitors LiCl and AR-A014418, as well as roscovitine, a cyclin-dependent kinase 5 inhibitor, decrease hypothermia-induced tau hyperphosphorylation, leading to different tau phosphorylation profiles. Therefore, we propose hypothermia-induced hyperphosphorylation as a reliable, fast, convenient and inexpensive tool to screen for tau kinase inhibitors.


Asunto(s)
Evaluación Preclínica de Medicamentos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Anestesia , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Hipotermia/inducido químicamente , Hipotermia/enzimología , Cloruro de Litio/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...