Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Aging (Albany NY) ; 12(13): 13422-13436, 2020 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-32652518

RESUMEN

Codonopsis pilosula Polysaccharides (CPPs), a traditional Chinese medicine used for thousands of years, is a potential neuroprotective polysaccharide via a relatively poorly understood mechanism. We previously reported that CPPs attenuated tau pathology in hTau transfected mice and therefore in the current work investigated the effect of CPPs on Aß toxicity and cognitive defects in APP/PS1 mice model. It was found that one-month intragastric administration of CPPs significantly ameliorated cognitive defects in APP/PS1 mice. In addition, CPPs treatment mitigated the loss of the synaptic plasticity and increased the synaptic proteins including synaptotagmin and PSD95. The expression of Aß42 and Aß40 was remarkably decreased in the hippocampus of APP/PS1 mice after CPPs treatment. We also found that CPPs coincubation significantly reduced the amount of APPß and Aß42 expression in cells. Intriguingly, the activity of BACE1 was decreased following CPPs treatment in both the hippocampus of APP/PS1 mice and in vitro experiments. Collectively, these results indicated that CPPs attenuated Aß pathology in APP/PS1 mice, and down-regulating BACE1 might be the underlaying mechanism which could be a therapeutic target for alleviating cognitive defects in AD pathology.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Codonopsis/química , Disfunción Cognitiva/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Polisacáridos/farmacología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Polisacáridos/uso terapéutico , Presenilina-1/genética
2.
Transl Neurodegener ; 9: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32266063

RESUMEN

Background: The homeostasis of metal ions, such as iron, copper, zinc and calcium, in the brain is crucial for maintaining normal physiological functions. Studies have shown that imbalance of these metal ions in the brain is closely related to the onset and progression of Alzheimer's disease (AD), the most common neurodegenerative disorder in the elderly. Main body: Erroneous deposition/distribution of the metal ions in different brain regions induces oxidative stress. The metal ions imbalance and oxidative stress together or independently promote amyloid-ß (Aß) overproduction by activating ß- or γ-secretases and inhibiting α-secretase, it also causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3ß (GSK-3ß), cyclin-dependent protein kinase-5 (CDK5), mitogen-activated protein kinases (MAPKs), etc., and inhibiting protein phosphatase 2A (PP2A). The metal ions imbalances can also directly or indirectly disrupt organelles, causing endoplasmic reticulum (ER) stress; mitochondrial and autophagic dysfunctions, which can cause or aggravate Aß and tau aggregation/accumulation, and impair synaptic functions. Even worse, the metal ions imbalance-induced alterations can reversely exacerbate metal ions misdistribution and deposition. The vicious cycles between metal ions imbalances and Aß/tau abnormalities will eventually lead to a chronic neurodegeneration and cognitive deficits, such as seen in AD patients. Conclusion: The metal ions imbalance induces Aß and tau pathologies by directly or indirectly affecting multiple cellular/subcellular pathways, and the disrupted homeostasis can reversely aggravate the abnormalities of metal ions transportation/deposition. Therefore, adjusting metal balance by supplementing or chelating the metal ions may be potential in ameliorating AD pathologies, which provides new research directions for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Metales/metabolismo , Péptidos beta-Amiloides/metabolismo , Química Encefálica , Humanos , Iones , Proteínas tau/metabolismo
3.
Neurosci Bull ; 35(2): 325-335, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30264378

RESUMEN

Hyperhomocysteinemia (Hhcy) is an independent risk factor for Alzheimer's disease (AD). Visual dysfunction is commonly found and is positively correlated with the severity of cognitive defects in AD patients. Our previous study demonstrated that Hhcy induces memory deficits with AD-like tau and amyloid-ß (Aß) pathologies in the hippocampus, and supplementation with folate and vitamin B12 (FB) prevents the Hhcy-induced AD-like pathologies in the hippocampus. Here, we investigated whether Hhcy also induces AD-like pathologies in the retina and the effects of FB. An Hhcy rat model was produced by vena caudalis injection of homocysteine for 14 days, and the effects of FB were assessed by simultaneous supplementation with FB in drinking water. We found that Hhcy induced vessel damage with Aß and tau pathologies in the retina, while simultaneous supplementation with FB remarkably attenuated the Hhcy-induced tau hyperphosphorylation at multiple AD-related sites and Aß accumulation in the retina. The mechanisms involved downregulation of amyloid precursor protein (APP), presenilin-1, beta-site APP-cleaving enzyme 1, and protein phosphatase-2A. Our data suggest that the retina may serve as a window for evaluating the effects of FB on hyperhomocysteinemia-induced Alzheimer-like pathologies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Ácido Fólico/uso terapéutico , Hiperhomocisteinemia/terapia , Retina/patología , Vitamina B 12/uso terapéutico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Homocisteína , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Masculino , Ratas Sprague-Dawley , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Proteínas tau/metabolismo
4.
Front Mol Neurosci ; 11: 437, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30542264

RESUMEN

Codonopsis pilosula polysaccharide (CPPs), a natural products with potentially lower toxicity and better bioavailability has been used in traditional Chinese medicine for 1000s of years and a neuroprotective polysaccharide mitigates tau pathology in Alzheimer's disease (AD) mouse model. However, whether CPPs can relieve AD pathology and cognitive defects remains poorly understood. Here we reported that CPPs remarkably increased the cell viability and PP2A activity, decreased tau phosphorylation in HEK 293/tau cells. Next, we employed an adeno-associated virus serotype 2 (AAV2)-induced expression of human full length tau (hTau) in C57/BL6 mice to mimic AD tau pathology. One month intragastric administration of CPPs significantly increased PP2A activity and reduced tau phosphorylation at Ser199, Ser202/Thr205 (AT8) and Thr231 in hippocampus of AAV2-hTau infected mice. Furthermore, behavioral tests revealed that CPPs rescued hTau overexpression induced cognitive defects while CPPs significantly increased the fEPSP slope and synaptic proteins including synaptotagmin and synaptophysin. Together, our data suggest that CPPs might prevent AD-like tau hyperphosphorylation via activation of PP2A and attenuates AD-like cognitive impairments through restoring the synaptic plasticity and synaptogenesis. In conclusion, our findings suggest that CPPs might be a potential candidate compound for the treatment of tau related diseases.

5.
Aging (Albany NY) ; 10(11): 3229-3248, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30425189

RESUMEN

Although few drugs are available today for the management of Alzheimer's disease (AD) and many plants and their extracts are extensively employed in animals' studies and AD patients, yet no drug or plant extract is able to reverse AD symptoms adequately. In the present study, Tamarix gallica (TG), a naturally occurring plant known for its strong antioxidative, anti-inflammatory and anti-amyloidogenic properties, was evaluated on homocysteine (Hcy) induced AD-like pathology and cognitive impairments in rats. We found that TG attenuated Hcy-induced oxidative stress and memory deficits. TG also improved neurodegeneration and neuroinflammation by upregulating synaptic proteins such as PSD95 and synapsin 1 and downregulating inflammatory markers including CD68 and GFAP with concomitant decrease in proinflammatory mediators interlukin-1ß (IL1ß) and tumor necrosis factor α (TNFα). TG attenuated tau hyperphosphorylation at multiple AD-related sites through decreasing some kinases and increasing phosphatase activities. Moreover, TG rescued amyloid-ß (Aß) pathology through downregulating BACE1. Our data for the first time provide evidence that TG attenuates Hcy-induced AD-like pathological changes and cognitive impairments, making TG a promising candidate for the treatment of AD-associated pathological changes.


Asunto(s)
Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Homocisteína/toxicidad , Hiperhomocisteinemia/inducido químicamente , Extractos Vegetales/farmacología , Tamaricaceae/química , Animales , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Metanol , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Compuestos de Fósforo , Fosfotransferasas/metabolismo , Fitoterapia , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley , Proteínas tau/metabolismo
6.
J Alzheimers Dis ; 63(3): 1141-1159, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29710724

RESUMEN

Alzheimer's disease (AD) is multifactorial with unclear etiopathology. Due to the complexity of AD, many attempted single therapy treatments, like Aß immunization, have generally failed. Therefore, there is a need for drugs with multiple benefits. Naturally occurring phytochemicals with neuroprotective, anti-amyloidogenic, antioxidative, and anti-inflammatory properties could be a possible way out. In this study, the effect of Moringa oleifera (MO), a naturally occurring plant with high antioxidative, anti-inflammatory, and neuroprotective effects, was evaluated on hyperhomocysteinemia (HHcy) induced AD-like pathology in rats. Homocysteine (Hcy) injection for 14 days was used to induce AD-like pathology. Simultaneous MO extract gavage followed the injection as a preventive treatment or, after injection completion, MO gavage was performed for another 14 days as a curative treatment. MO was found to not only prevent but also rescue the oxidative stress and cognitive impairments induced by Hcy treatment. Moreover, MO recovered the decreased synaptic proteins PSD93, PSD95, Synapsin 1 and Synaptophysin, and improved neurodegeneration. Interestingly, MO decreased the Hyc-induced tau hyperphosphorylation at different sites including S-199, T-231, S-396, and S-404, and at the same time decreased Aß production through downregulation of BACE1. These effects in HHcy rats were accompanied by a decrease in calpain activity under MO treatment, supporting that calpain activation might be involved in AD pathogenesis in HHcy rats. Taken together, our data, for the first time, provided evidence that MO alleviates tau hyperphosphorylation and Aß pathology in a HHcy AD rat model. This and previous other studies support MO as a good candidate for, and could provide new insights into, the treatment of AD and other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Trastornos del Conocimiento , Homocisteína/toxicidad , Moringa oleifera , Extractos Vegetales/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata , Proteínas tau/metabolismo
7.
Curr Alzheimer Res ; 15(1): 89-99, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28847282

RESUMEN

BACKGROUND: Ginkgo biloba extract EGb761 has shown the neuroprotective effects on Alzheimer's disease (AD) through the protection against the Aß-induced neurotoxicity. However, it is not completedly clear whether EGb761 attenuates tau hyperphosphorylation, another of the most prominent mechanisms underlying the pathology of AD. METHODS: we employed hyperhomocysteinemia (HHcy) to mimic AD like pathological alterations and memory deficits in rats as model, and injected EGb761 with or after HHcy injection as prevention and treatment, injected saline as control. We measured the status of oxidative damage and spatial and learning memory in rats. Then we detected the level of memory-related proteins, tau phosphorylation and the level and activity of tau kinase (GSK-3ß) and phosphatase (PP2A) by Western blotting and Immunohistochemistry. RESULTS: We found that EGb761 could significantly antagonize HHcy-induced oxidative damage, recover PP2Ac and GSK3ß activities deregulated by HHcy. Furthermore, tau was hyperphosphorylated at Thr231, Ser262, Ser396, and Ser404, most common PP2Ac and GSK3ß targeted sites in the hippocampus and prefrontal cortex of HHcy rats, whereas EGb761 recovered the tau phosphorylation at those sites. Behavioral tests revealed that EGb761 rescued HHcy-induced spatial reference memory deficit and upregulated the expression of synapse-associated protein PSD95 and synapsin-1. CONCLUSION: EGb761 might be a promising drug to treat AD through its anti-oxidative activity and decreasing tau hyperphosphorylation besides the protection against the Aß-induced neurotoxicity.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Hiperhomocisteinemia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Extractos Vegetales/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Ginkgo biloba , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Hiperhomocisteinemia/psicología , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Sinapsinas/metabolismo
8.
J Cell Mol Med ; 21(12): 3515-3528, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28656644

RESUMEN

The cholinergic impairment is an early marker in Alzheimer's disease (AD), while the mechanisms are not fully understood. We investigated here the effects of glycogen synthase kinse-3 (GSK-3) activation on the cholinergic homoeostasis in nucleus basalis of Meynert (NBM) and frontal cortex, the cholinergic enriched regions. We activated GSK-3 by lateral ventricular infusion of wortmannin (WT) and GF-109203X (GFX), the inhibitors of phosphoinositol-3 kinase (PI3-K) and protein kinase C (PKC), respectively, and significantly decreased the acetylcholine (ACh) level via inhibiting choline acetyl transferase (ChAT) rather than regulating acetylcholinesterase (AChE). Neuronal axonal transport was disrupted and ChAT accumulation occurred in NBM and frontal cortex accompanied with hyperphosphorylation of tau and neurofilaments. Moreover, ChAT expression decreased in NBM attributing to cleavage of nuclear factor-κB/p100 into p52 for translocation into nucleus to lower ChAT mRNA level. The cholinergic dysfunction could be mimicked by overexpression of GSK-3 and rescued by simultaneous administration of LiCl or SB216763, inhibitors of GSK-3. Our data reveal the molecular mechanism that may underlie the cholinergic impairments in AD patients.


Asunto(s)
Acetilcolina/metabolismo , Núcleo Basal de Meynert/metabolismo , Lóbulo Frontal/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Acetilcolinesterasa/genética , Acetilcolinesterasa/metabolismo , Androstadienos/farmacología , Animales , Transporte Axonal/efectos de los fármacos , Núcleo Basal de Meynert/efectos de los fármacos , Núcleo Basal de Meynert/patología , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/patología , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Indoles/farmacología , Cloruro de Litio/farmacología , Masculino , Maleimidas/farmacología , FN-kappa B/genética , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Técnicas Estereotáxicas , Wortmanina , Proteínas tau/genética , Proteínas tau/metabolismo
9.
J Alzheimers Dis ; 42(3): 1029-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25024319

RESUMEN

Alzheimer's disease (AD) has multiple etiopathogenic factors, yet the definitive cause remains unclear and the therapeutic strategies have been elusive. Combination therapy, as one of the promising treatments, has been studied for years and may exert synergistic beneficial effects on AD through polytherapeutic targets. In this study, we tested the effects of a synthesized juxtaposition (named SCR1693) composed of an acetylcholinesterase inhibitor (AChEI) and a calcium channel blocker (CCB) on the hyperhomocysteinemia (HHcy)-induced AD rat model, and found that SCR1693 remarkably improved the HHcy-induced memory deficits and preserved dendrite morphologies as well as spine density by upregulating synapse-associated proteins PSD95 and synapsin-1. In addition, SCR1693 attenuated HHcy-induced tau hyperphosphorylation at multiple AD-associated sites by regulating the activity of protein phosphatase-2A and glycogen synthase kinase-3ß. Furthermore, SCR1693 was more effective than individual administration of both donepezil and nilvadipine which were used as AChEI and CCB, respectively, in the clinical practice. In conclusion, our data suggest that the polytherapeutic targeting juxtaposition SCR1693 (AChEI-CCB) is a promising therapeutic candidate for AD.


Asunto(s)
Encefalopatías , Bloqueadores de los Canales de Calcio/uso terapéutico , Inhibidores de la Colinesterasa/uso terapéutico , Hiperhomocisteinemia/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Animales , Encefalopatías/tratamiento farmacológico , Encefalopatías/etiología , Encefalopatías/patología , Modelos Animales de Enfermedad , Donepezilo , Quimioterapia Combinada , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/ultraestructura , Homocisteína/toxicidad , Hiperhomocisteinemia/inducido químicamente , Indanos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Nifedipino/análogos & derivados , Nifedipino/uso terapéutico , Piperidinas/uso terapéutico , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Corteza Prefrontal/ultraestructura , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata , Tacrina/análogos & derivados , Tacrina/farmacología , Tacrina/uso terapéutico
10.
Neuropharmacology ; 76 Pt A: 80-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24012657

RESUMEN

Alzheimer's disease (AD) is characterized by the cholinergic neurons loss and impairments of learning and memory. Scopolamine is common used to imitate AD pathological features and also causes an obvious oxidative stress. In this study, we found that intraperitoneal administration of supplementary acetyl-L-carnitine partially reverses the learning and memory defects induced by scopolamine. We also found that acetyl-L-carnitine reverses the impairment of long-term potentiation, dendritic abnormalities, and the impaired recruitment of synaptic protein. The beneficial effects of acetyl-L-carnitine may occur through amelioration of oxidative stress because it effectively decreases the levels of oxidative products and increases the activity of superoxide dismutase; this leads to a recovery in the suppressed activity of p53 caused oxidative stimuli, which in turn restores levels of insulin-like growth factor II, an important hormone for learning and memory. Our study provides the first evidence of the potential utility of acetyl-L-carnitine in treating the synaptic disorders prevalent in AD and other neurodegenerative diseases. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.


Asunto(s)
Acetilcarnitina/farmacología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Oxidación-Reducción/efectos de los fármacos , Escopolamina , Proteína p53 Supresora de Tumor/metabolismo , Acetilcarnitina/uso terapéutico , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Ratas , Superóxido Dismutasa/metabolismo
11.
Int J Mol Sci ; 14(7): 14575-93, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23857055

RESUMEN

Alzheimer's disease (AD), an age-related neurodegenerative disorder with progressive cognition deficit, is characterized by extracellular senile plaques (SP) of aggregated ß-amyloid (Aß) and intracellular neurofibrillary tangles, mainly containing the hyperphosphorylated microtubule-associated protein tau. Multiple factors contribute to the etiology of AD in terms of initiation and progression. Melatonin is an endogenously produced hormone in the brain and decreases during aging and in patients with AD. Data from clinical trials indicate that melatonin supplementation improves sleep, ameliorates sundowning and slows down the progression of cognitive impairment in AD patients. Melatonin efficiently protects neuronal cells from Aß-mediated toxicity via antioxidant and anti-amyloid properties. It not only inhibits Aß generation, but also arrests the formation of amyloid fibrils by a structure-dependent interaction with Aß. Our studies have demonstrated that melatonin efficiently attenuates Alzheimer-like tau hyperphosphorylation. Although the exact mechanism is still not fully understood, a direct regulatory influence of melatonin on the activities of protein kinases and protein phosphatases is proposed. Additionally, melatonin also plays a role in protecting the cholinergic system and in anti-inflammation. The aim of this review is to stimulate interest in melatonin as a potentially useful agent in the prevention and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Melatonina/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Humanos , Fosforilación , Proteínas tau/metabolismo
12.
PLoS One ; 8(1): e55384, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383172

RESUMEN

Zinc ions highly concentrate in hippocampus and play a key role in modulating spatial learning and memory. At a time when dietary fortification and supplementation of zinc have increased the zinc consuming level especially in the youth, the toxicity of zinc overdose on brain function was underestimated. In the present study, weaning ICR mice were given water supplemented with 15 ppm Zn (low dose), 60 ppm Zn (high dose) or normal lab water for 3 months, the behavior and brain zinc homeostasis were tested. Mice fed high dose of zinc showed hippocampus-dependent memory impairment. Unexpectedly, zinc deficiency, but not zinc overload was observed in hippocampus, especially in the mossy fiber-CA3 pyramid synapse. The expression levels of learning and memory related receptors and synaptic proteins such as NMDA-NR2A, NR2B, AMPA-GluR1, PSD-93 and PSD-95 were significantly decreased in hippocampus, with significant loss of dendritic spines. In keeping with these findings, high dose intake of zinc resulted in decreased hippocampal BDNF level and TrkB neurotrophic signaling. At last, increasing the brain zinc level directly by brain zinc injection induced BDNF expression, which was reversed by zinc chelating in vivo. These results indicate that zinc plays an important role in hippocampus-dependent learning and memory and BDNF expression, high dose supplementation of zinc induces specific zinc deficiency in hippocampus, which further impair learning and memory due to decreased availability of synaptic zinc and BDNF deficit.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Trastornos de la Memoria/inducido químicamente , Transducción de Señal/efectos de los fármacos , Zinc/deficiencia , Zinc/toxicidad , Análisis de Varianza , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Suplementos Dietéticos , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Guanilato-Quinasas/metabolismo , Hipocampo/metabolismo , Técnicas Histológicas , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Receptor trkB/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Zinc/administración & dosificación
13.
J Neurochem ; 124(3): 388-96, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23157378

RESUMEN

Hyperhomocysteinemia (Hhcy) may induce memory deficits with ß-amyloid (Aß) accumulation and tau hyperphosphorylation. Simultaneous supplement of folate and vitamin B12 partially restored the plasma homocysteine level and attenuated tau hyperphosphorylation, Aß accumulation and memory impairments induced by Hhcy. However, folate and vitamin B12 treatment have no effects on Hhcy which has the methylenetetrahydrofolate reductase genotype mutation. In this study, we investigated the effects of simultaneous supplement of betaine on Alzheimer-like pathological changes and memory deficits in hyperhomocysteinemic rats after a 2-week induction by vena caudalis injection of homocysteine (Hcy). We found that supplementation of betaine could ameliorate the Hcy-induced memory deficits, enhance long-term potentiation (LTP) and increase dendritic branches numbers and the density of the dendritic spines, with up-regulation of NR1, NR2A, synaptotagmin, synaptophysin, and phosphorylated synapsin I protein levels. Supplementation of betaine also attenuated the Hcy-induced tau hyperphosphorylation at multiple AD-related sites through activation protein phosphatase-2A (PP2A) with decreased inhibitory demethylated PP2A(C) at Leu309 and phosphorylated PP2A(C) at Tyr307. In addition, supplementation of betaine also decreased Aß production with decreased presenilin-1 protein levels. Our data suggest that betaine could be a promising candidate for arresting Hcy-induced AD-like pathological changes and memory deficits.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Betaína/toxicidad , Homocisteína/toxicidad , Hiperhomocisteinemia/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Enfermedad de Alzheimer/sangre , Animales , Modelos Animales de Enfermedad , Homocisteína/sangre , Hiperhomocisteinemia/inducido químicamente , Lipotrópicos/farmacología , Masculino , Trastornos de la Memoria/inducido químicamente , Ratas , Ratas Sprague-Dawley
14.
Mol Neurobiol ; 47(1): 373-81, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23054680

RESUMEN

Alzheimer's disease (AD) is the most prevalent type of dementia in elderly people. There are decreased melatonin levels in the serum of AD patients, and melatonin supplements are able to reverse AD pathology and memory deficits in many animal experiments and clinical trials. However, the underlying mechanism regarding how melatonin rescues the AD-like memory/synaptic disorder remains unknown. Here, we use the Morris water maze, step-down inhibitory avoidance task, in vivo long-term potentiation recording, and Golgi staining and report that intraperitoneal injection of melatonin (1 mg/kg/day) for 14 days in rats effectively reverses the memory and synaptic impairment in scopolamine-induced amnesia, a well-recognized dementia animal model. Using real-time polymerase chain reaction and western blotting experiments, we further determined that melatonin rescues the EPACs/miR-124/Egr1 signal pathway, which is important in learning and memory, as reported recently. Our studies provide a novel underlying epigenetic mechanism for melatonin to attenuate the synaptic disorder and could benefit drug discovery in neurodegenerative diseases.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Melatonina/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , MicroARNs/metabolismo , Transducción de Señal , Sinapsis/patología , Animales , Masculino , Melatonina/farmacología , Trastornos de la Memoria/fisiopatología , Ratas , Ratas Sprague-Dawley , Escopolamina , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
15.
J Biol Chem ; 287(14): 11174-82, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334661

RESUMEN

Hyperphosphorylated tau is the major component of neurofibrillary tangles in Alzheimer disease (AD), and the tangle distribution largely overlaps with zinc-containing glutamatergic neurons, suggesting that zinc released in synaptic terminals may play a role in tau phosphorylation. To explore this possibility, we treated cultured hippocampal slices or primary neurons with glutamate or Bic/4-AP to increase the synaptic activity with or without pretreatment of zinc chelators, and then detected the phosphorylation levels of tau. We found that glutamate or Bic/4-AP treatment caused tau hyperphosphorylation at multiple AD-related sites, including Ser-396, Ser-404, Thr-231, and Thr-205, while application of intracellular or extracellular zinc chelators, or blockade of zinc release by extracellular calcium omission almost abolished the synaptic activity-associated tau hyperphosphorylation. The zinc release and translocation of excitatory synapses in the hippocampus were detected, and zinc-induced tau hyperphosphorylation was also observed in cultured brain slices incubated with exogenously supplemented zinc. Tau hyperphosphorylation induced by synaptic activity was strongly associated with inactivation of protein phosphatase 2A (PP2A), and this inactivation can be reversed by pretreatment of zinc chelator. Together, these results suggest that synaptically released zinc promotes tau hyperphosphorylation through PP2A inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Fosfatasa 2/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Zinc/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/efectos de los fármacos , Zinc/farmacología
16.
Rejuvenation Res ; 14(6): 669-79, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21978079

RESUMEN

Hyperhomocystinemia could induce tau protein hyperphosphorylation, ß-amyloid (Aß) accumulation, and memory deficits as seen in Alzheimer disease (AD), the most common cause of senile dementia with no effective cure currently. To search for possible treatment for AD, we produced a hyperhomocysteinemia model by vena caudalis injection of homocystine (Hcy) for 2 weeks and studied the effects of acetyl-L-carnitine (ALC) in rats. We found that simultaneous supplement of ALC could improve the Hcy-induced memory deficits remarkably, with attenuation of tau hyperphosphorylation and Aß accumulation. Supplement of ALC almost abolished the Hcy-induced tau hyperphosphorylation at multiple AD-related sites. Supplementation of ALC also suppressed the phosphorylation of ß-amyloid precursor proteins (APP), which may underlie the reduction of Aß. Our data suggest that ALC could be a promising candidate for arresting Hcy-induced AD-like pathological and behavioral impairments.


Asunto(s)
Acetilcarnitina/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Homocisteína/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Conducta Animal , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto , Fosforilación , Ratas , Ratas Sprague-Dawley , Tinción con Nitrato de Plata , Proteínas tau/metabolismo
17.
J Alzheimers Dis ; 27(3): 639-50, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21860088

RESUMEN

Hyperhomocysteinemia is associated with an increased risk of Alzheimer's disease (AD). Our previous work has demonstrated that combined folate and vitamin B12 (vit-B12) supplementation prevents tau hyperphosphorylation and memory deficits induced by acute administration of homocysteine in young rats. Here, we further investigated whether folate/vit-B12 supplementation is also effective in aged rats with a chronically high level of homocysteine. 18-month-old rats were injected with homocysteine via the vena caudalis with or without a concurrent folate/vit-B12 supplementation for 28 weeks. We found that hyperhomocysteinemia induced tau hyperphosphorylation and accumulation in hippocampus and cortex. Concurrent signaling changes included the activation of glycogen synthase kinases-3ß, cyclin-dependent kinase-5, c-Jun N-terminal kinase, extracellular signal-regulated kinase, and p38MAPK, and inhibition of protein phosphatase 2A. Although the ability to learn was not affected, the aged rats exhibited significant memory deficits. Folate/vit-B12 supplementation attenuated these biochemical and behavioral correlates. These data demonstrate that folate/vit-B12 supplementation is also effective in a chronic hyperhomocysteinemia model in reversing the AD-like tau pathologies and memory deficits.


Asunto(s)
Envejecimiento/metabolismo , Ácido Fólico/administración & dosificación , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/prevención & control , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Vitamina B 12/administración & dosificación , Proteínas tau/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/psicología , Animales , Enfermedad Crónica , Suplementos Dietéticos , Modelos Animales de Enfermedad , Quimioterapia Combinada , Masculino , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas tau/antagonistas & inhibidores
18.
Am J Pathol ; 174(4): 1481-91, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19264913

RESUMEN

Hyperhomocysteinemia and beta-amyloid (Abeta) overproduction are critical etiological and pathological factors in Alzheimer disease, respectively; however, the intrinsic link between them is still missing. Here, we found that Abeta levels increased and amyloid precursor protein (APP) levels simultaneously decreased in hyperhomocysteinemic rats after a 2-week induction by vena caudalis injection of homocysteine. Concurrently, both the mRNA and protein levels of presenilin-1, a component of gamma-secretase, were elevated, whereas the expression levels of beta-secretase and presenilin-2 were not altered. We also observed that levels of phosphorylated APP at threonine-668, a crucial site facilitating the amyloidogenic cleavage of APP, increased in rats with hyperhomocysteinemia, although the phosphorylation per se did not increase the binding capacity of pT668-APP to the secretases. The enhanced phosphorylation of APP in these rats was not relevant to either c-Jun N-terminal kinase or cyclin-dependent kinase-5. A prominent spatial memory deficit was detected in rats with hyperhomocysteinemia. Simultaneous supplementation of folate and vitamin-B12 attenuated the hyperhomocysteinemia-induced abnormal processing of APP and improved memory. Our data revealed that hyperhomocysteinemia could increase Abeta production through the enhanced expression of gamma-secretase and APP phosphorylation, causing memory deficits that could be rescued by folate and vitamin-B12 treatment in these rats. It is suggested that hyperhomocysteinemia may serve as an upstream factor for increased Abeta production as seen in patients with Alzheimer disease.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Expresión Génica , Hiperhomocisteinemia/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Ensayo de Inmunoadsorción Enzimática , Ácido Fólico/farmacología , Hiperhomocisteinemia/complicaciones , Inmunohistoquímica , Inmunoprecipitación , Trastornos de la Memoria/etiología , Fosforilación , Presenilina-1/metabolismo , Presenilina-2/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vitamina B 12/farmacología , Complejo Vitamínico B/farmacología
19.
J Alzheimers Dis ; 16(2): 287-300, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19221418

RESUMEN

Most patients with Alzheimer's disease (AD) present decreased levels of melatonin, a day-night rhythm-related hormone. To investigate the role of melatonin deficiency in AD, we used constant illumination to interrupt melatonin metabolism and measured some of the AD-like alterations in rats. Concomitant with decreased serum melatonin, the rats developed spatial memory deficits, tau hyperphosphorylation at multiple sites, activation of glycogen synthase kinase-3 and protein kinase A, as well as suppression of protein phosphatase-1. Prominent oxidative damage and organelle lesions, demonstrated by increased expression of endoplasmic reticulum (ER) stress-related proteins including BiP/GRP78 and CHOP/GADD153, decreased number of rough ER and free ribosome, thinner synapses, and increased superoxide dismutase and monoamine oxidase were also observed in the light exposed rats. Simultaneous supplement of melatonin partially arrested the behavioral and molecular impairments. It is suggested that melatonin deficiency may be an upstream effector responsible for the AD-like behavioral and molecular pathologies with ER stress-involved mechanisms.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Antioxidantes/uso terapéutico , Retículo Endoplásmico/patología , Melatonina/uso terapéutico , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/complicaciones , Animales , Conducta Animal , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/ultraestructura , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/ultraestructura , Iluminación/efectos adversos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Melatonina/sangre , Microscopía Inmunoelectrónica/métodos , Monoaminooxidasa/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/efectos de la radiación , Superóxido Dismutasa/metabolismo , Proteínas tau/metabolismo
20.
Neurobiol Aging ; 29(11): 1654-65, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17537547

RESUMEN

Hyperhomocysteinemia increases the risk of Alzheimer's disease (AD), but the mechanism is elusive. Here, we found that high plasma homocysteine induced by vena caudalis injection for 2 weeks could induce AD-like tau hyperphosphorylation at multiple sites in rat brain hippocampus. Homocysteine inhibited the activity of protein phosphatase 2A (PP2A) with a simultaneously increased Leu(309)-demethylation and Tyr(307)-phosphorylation of PP2A catalytic subunit (PP2A(C)). PP2A(C) Leu(309)-demethylation was positively correlated with its Tyr(307)-phosphorylation; and the abnormally modified PP2A(C) was incompetent in binding to its regulatory subunit (PP2A(B)). Homocysteine also activated methylesterase which stimulates demethylation of PP2A(C). In hippocampal slices of the homocysteine injected-rats and of the AD patients, the demethylated but not the methylated PP2A(C) was co-localized with the hyperphosphorylated tau. A simultaneous supplement of folate and vitamin B12 restored partially the plasma homocysteine level and thus significantly antagonized the homocysteine-induced tau hyperphosphorylation and as well as PP2A inactivation and the activity-related modifications of PP2A(C). These results suggest that homocysteine may be an upstream effector to induce AD-like tau hyperphosphorylation through inactivating PP2A.


Asunto(s)
Hipocampo/metabolismo , Homocisteína/administración & dosificación , Homocisteína/sangre , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Transducción de Señal/fisiología , Proteínas tau/metabolismo , Animales , Activación Enzimática , Hipocampo/efectos de los fármacos , Inyecciones Intravenosas , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA