Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Ther ; 21(12): 2247-57, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23922015

RESUMEN

Deficits of protein phosphatase-2A (PP2A) play a crucial role in tau hyperphosphorylation, amyloid overproduction, and synaptic suppression of Alzheimer's disease (AD), in which PP2A is inactivated by the endogenously increased inhibitory protein, namely inhibitor-2 of PP2A (I2(PP2A)). Therefore, in vivo silencing I2(PP2A) may rescue PP2A and mitigate AD neurodegeneration. By infusion of lentivirus-shRNA targeting I2(PP2A) (LV-siI2(PP2A)) into hippocampus and frontal cortex of 11-month-old tg2576 mice, we demonstrated that expression of LV-siI2(PP2A) decreased remarkably the elevated I2(PP2A) in both mRNA and protein levels. Simultaneously, the PP2A activity was restored with the mechanisms involving reduction of the inhibitory binding of I2(PP2A) to PP2A catalytic subunit (PP2AC), repression of the inhibitory Leu309-demethylation and elevation of PP2AC. Silencing I2(PP2A) induced a long-lasting attenuation of amyloidogenesis in tg2576 mice with inhibition of amyloid precursor protein hyperphosphorylation and ß-secretase activity, whereas simultaneous inhibition of PP2A abolished the antiamyloidogenic effects of I2(PP2A) silencing. Finally, silencing I2(PP2A) could improve learning and memory of tg2576 mice with preservation of several memory-associated components. Our data reveal that targeting I2(PP2A) can efficiently rescue Aß toxicities and improve the memory deficits in tg2576 mice, suggesting that I2(PP2A) could be a promising target for potential AD therapies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Lentivirus/genética , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/genética , Proteína Fosfatasa 2/metabolismo , Interferencia de ARN , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Dominio Catalítico , Línea Celular Tumoral , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Vectores Genéticos , Células HEK293 , Hipocampo/metabolismo , Chaperonas de Histonas , Humanos , Lentivirus/metabolismo , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Proteína Fosfatasa 2/química , ARN Interferente Pequeño/genética
2.
J Huazhong Univ Sci Technolog Med Sci ; 33(3): 368-374, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23771662

RESUMEN

This study investigated the effects of benazepril administered in the morning or evening on the diurnal variation of renin-angiotensin-aldosterone system (RAAS) and clock genes in the kidney. The male Wistar rat models of 5/6 subtotal nephrectomy (STNx) were established. Animals were randomly divided into 4 groups: sham STNx group (control), STNx group, morning benazepril group (MB) and evening benazepril group (EB). Benazepril was intragastrically administered at a dose of 10 mg/kg/day at 07:00 and 19:00 in the MB group and EB group respectively for 12 weeks. All the animals were synchronized to the light:dark cycle of 12:12 for 12 weeks. Systolic blood pressure (SBP), 24-h urinary protein excretion and renal function were measured at 11 weeks. Blood samples and kidneys were collected every 4 h throughout a day to detect the expression pattern of renin activity (RA), angiotensin II (AngII) and aldosterone (Ald) by radioimmunoassay (RIA) and the mRNA expression profile of clock genes (bmal1, dbp and per2) by real-time PCR at 12 weeks. Our results showed that no significant differences were noted in the SBP, 24-h urine protein excretion and renal function between the MB and EB groups. There were no significant differences in average Ald and RA content of a day between the MB group and EB group. The expression peak of bmal1 mRNA was phase-delayed by 4 to 8 h, and the diurnal variation of per2 and dbp mRNA diminished in the MB and EB groups compared with the control and STNx groups. It was concluded when the similar SBP reduction, RAAS inhibition and clock gene profile were achieved with optimal dose of benazepril, morning versus evening dosing of benazepril has the same renoprotection effects.


Asunto(s)
Benzazepinas/administración & dosificación , Proteínas CLOCK/metabolismo , Hipertensión Renal/tratamiento farmacológico , Hipertensión Renal/fisiopatología , Riñón/efectos de los fármacos , Riñón/fisiopatología , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Antihipertensivos/administración & dosificación , Ritmo Circadiano , Cronoterapia de Medicamentos , Perfilación de la Expresión Génica , Riñón/cirugía , Masculino , Nefrectomía , Ratas , Ratas Wistar , Resultado del Tratamiento
3.
J Neurosci ; 30(10): 3839-48, 2010 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-20220019

RESUMEN

Protein phosphatase 2A (PP2A) is indispensable in development, and deficits of PP2A and deterioration of neuronal axons have been observed in several neurodegenerative disorders, but the direct link between PP2A and the neuronal axon development is still missing. Here, we show that PP2A is essential for axon development in transfected rat brain and the dissociated hippocampal neurons. Upregulation of PP2A catalytic subunit (PP2Ac) not only promotes formation and elongation of the functional axons but also rescues axon retardation induced by PP2A inhibition. PP2A can dephosphorylate collapsin response mediator protein-2 (CRMP2) that implements the axon polarization, whereas constitutive expression of phosphomimic-CRMP2 abrogates the effect of PP2A upregulation. We also demonstrate that PP2Ac is enriched in the distal axon of the hippocampal neurons. Our results reveal a mechanistic link between PP2A and axonogenesis/axonopathy, suggesting that upregulation of PP2A may be a promising therapeutic for some neurodegenerative disorders.


Asunto(s)
Axones/enzimología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Proteína Fosfatasa 2/fisiología , Sustitución de Aminoácidos/genética , Animales , Axones/metabolismo , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Hipocampo/citología , Hipocampo/enzimología , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuritas/enzimología , Neuritas/metabolismo , Neurogénesis/genética , Fosforilación/genética , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/biosíntesis , Proteína Fosfatasa 2/genética , ARN Interferente Pequeño/fisiología , Ratas
4.
Neurochem Res ; 36(2): 288-96, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21061060

RESUMEN

Altered neurogenesis has been reported in Alzheimer disease (AD), the most common neurodegenerative disorder characterized with hyperphosphorylated tau and accumulation of ß-amyloid (Aß). Recent studies suggest that tau phosphorylation is essential for hippocampal neurogenesis, however, it is not known whether tau phosphorylation also play a role in neurogenesis of subventricular zone (SVZ), another main progenitor niche in the brain. Here, we examined the expression of phosphorylated tau (p-tau) in SVZ and analyzed the role of p-tau in adult SVZ neurogenesis. We found that the expression of p-tau increased during postnatal development and remains at a high level until adulthood, and the p-tau was colocalized with some SVZ neural precursors. However, up-regulating glycogen synthase kinase-3 (GSK-3), a crucial tau kinase, had no effect on SVZ neurogenesis in adult rat brain. The SVZ neurogenesis was also unaffected in tau knockout and human tau transgenic mice. These results suggest that tau phosphorylation and GSK-3 activation may not be essential for adult SVZ neurogenesis.


Asunto(s)
Encéfalo/anatomía & histología , Glucógeno Sintasa Quinasa 3/metabolismo , Neurogénesis/fisiología , Nicho de Células Madre , Proteínas tau/metabolismo , Animales , Biomarcadores/metabolismo , Encéfalo/fisiología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas tau/genética
5.
Hippocampus ; 20(12): 1339-49, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19816983

RESUMEN

An increased hippocampal neurogenesis has been observed in Alzheimer disease (AD), the most common neurodegenerative disorder characterized with accumulation of ß-amyloid (Aß) and hyperphosphorylated tau (p-tau). Studies in transgenic mouse models suggest that the amyloidosis suppresses adult neurogenesis. Although emerging evidence links tau to neurodevelopment, the direct data regarding tau phosphorylation in adult neurogenesis is missing. Here, we found that the immature neurons, identified by doublecortin (DCX) and neurogenic differentiation factor (neuroD), were only immunoreactive to p-tau but not to the non-p-tau in adult rat brain and human patients with AD, and the p-tau was coexpressed temporally and spatially with DCX and neuroD in the hippocampal dentate gyrus (DG) of the rat brains during postnatal development. A correlative increase of immature neuron markers and tau phosphorylation was induced in rat hippocampal DG by upregulating glycogen synthase kinase-3 (GSK-3), a crucial tau kinase, and the increased neurogenesis was due to an enhanced proliferation but not survival or differentiation of the newborn neurons. The hippocampal neurogenesis was severely impaired in tau knockout mice and activation of GSK-3 in these mice did not rescue the deficits. These results reveal an essential role of tau phosphorylation in adult hippocampal neurogenesis. It suggests that spatial/temporal manipulation of tau phosphorylation may be compensatory for the neuron loss in neurological disorders, including AD.


Asunto(s)
Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Proteínas tau/metabolismo , Análisis de Varianza , Animales , Western Blotting , Proteína Doblecortina , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Fosforilación , Ratas , Ratas Sprague-Dawley
6.
Pharmacol Biochem Behav ; 117: 47-51, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24342459

RESUMEN

Synaptic communication forms the basis of learning and memory. Disruptions of synaptic function and memory have been widely reported in many neurological diseases, such as dementia. Thus, restoration of impaired synaptic communication is a potential therapeutic approach for these diseases. In this study, we demonstrated that supplementation with berberine, a plant alkaloid with a long history of medicinal usage in Chinese medicine, effectively reverses the synaptic deficits induced by D-galactose. We also found that berberine rescued D-galactose-induced memory impairment and additionally rescued the mRNA and protein levels of Arc/Arg3.1, an important immediate early gene that is crucial for maintaining normal synaptic plasticity. Our study provides the first piece of evidence supporting the potential use of berberine in the treatment of neural diseases with synaptic/memory impairments.


Asunto(s)
Berberina/farmacología , Proteínas del Citoesqueleto/metabolismo , Galactosa/farmacología , Trastornos de la Memoria/prevención & control , Proteínas del Tejido Nervioso/metabolismo , Sinapsis/efectos de los fármacos , Animales , Secuencia de Bases , Cartilla de ADN , Masculino , Trastornos de la Memoria/inducido químicamente , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Neurobiol Aging ; 34(6): 1555-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23402899

RESUMEN

The current therapies for Alzheimer's disease (AD) are merely palliative that cannot arrest the pathologic progression of the disease. Therefore, it is critical to develop treatments that can target the disease-modifying molecule(s). In the present study, we found that treatment of tg2576 mice with melatonin from 4-8 months of age did not improve the pathology or behavioral performance of the mice. However, remarkable attenuation of tau and ß-amyloid pathologies with memory improvement were observed when melatonin was supplied from the age of 8-12 months or 4-12 months of the mice; more importantly, the improvements were still significant when the mice survived to old age. We also found that the disease stage-specific alteration of glycogen synthase kinase-3ß (GSK-3ß) but not protein phosphatase-2A, was correlated with the alterations of the pathology and behavior, and the timely targeting of GSK-3ß was critical for the efficacy of melatonin. Our finding suggests that melatonin treatment only at proper timing could arrest AD by targeting the activated GSK-3ß, which provides primary evidence for the importance and strategy in developing disease-modifying interventions of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Melatonina/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Animales , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Melatonina/farmacología , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/patología , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
8.
Neurobiol Aging ; 33(2): 254-64, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20138402

RESUMEN

A chronic neuron loss is the cardinal pathology in Alzheimer disease (AD), but it is still not understood why most neurons in AD brain do not accomplish apoptosis even though they are actually exposed to an environment with enriched proapoptotic factors. Protein phosphatase-2A inhibitor-2 (I(2)(PP2A)), an endogenous PP2A inhibitor, is significantly increased in AD brain, but the role of I(2)(PP2A) in AD-like neuron loss is elusive. Here, we show that I(2)(PP2A) regulates p53 and Akt correlatively. The mechanisms involve activated transcription and p38 MAPK activities. More importantly, we demonstrate that the simultaneous activation of Akt induced by I(2)(PP2A) counteracts the hyperactivated p53-induced cell apoptosis. Furthermore, I(2)(PP2A), p53 and Akt are all elevated in the brain of mouse model and AD patients. Our results suggest that the increased I(2)(PP2A) may trigger apoptosis by p53 upregulation, but due to simultaneous activation of Akt, the neurons are aborted from the apoptotic pathway. This finding contributes to the understanding of why most neurons in AD brain do not undergo apoptosis.


Asunto(s)
Apoptosis/fisiología , Chaperonas de Histonas/metabolismo , Neuronas/citología , Neuronas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Unión al ADN , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA