Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Neuroinflammation ; 18(1): 146, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183019

RESUMEN

BACKGROUND: Thymosin ß4 (Tß4) is the most abundant member of the ß-thymosins and plays an important role in the control of actin polymerization in eukaryotic cells. While its effects in multiple organs and diseases are being widely investigated, the safety profile has been established in animals and humans, currently, little is known about its influence on Alzheimer's disease (AD) and the possible mechanisms. Thus, we aimed to evaluate the effects and mechanisms of Tß4 on glial polarization and cognitive performance in APP/PS1 transgenic mice. METHODS: Behavior tests were conducted to assess the learning and memory, anxiety and depression in APP/PS1 mice. Thioflavin S staining, Nissl staining, immunohistochemistry/immunofluorescence, ELISA, qRT-PCR, and immunoblotting were performed to explore Aß accumulation, phenotypic polarization of glial cells, neuronal loss and function, and TLR4/NF-κB axis in APP/PS1 mice. RESULTS: We demonstrated that Tß4 protein level elevated in all APP/PS1 mice. Over-expression of Tß4 alone alleviated AD-like phenotypes of APP/PS1 mice, showed less brain Aß accumulation and more Insulin-degrading enzyme (IDE), reversed phenotypic polarization of microglia and astrocyte to a healthy state, improved neuronal function and cognitive behavior performance, and accidentally displayed antidepressant-like effect. Besides, Tß4 could downregulate both TLR4/MyD88/NF-κB p65 and p52-dependent inflammatory pathways in the APP/PS1 mice. While combination drug of TLR4 antagonist TAK242 or NF-κB p65 inhibitor PDTC exerted no further effects. CONCLUSIONS: These results suggest that Tß4 may exert its function by regulating both classical and non-canonical NF-κB signaling and is restoring its function as a potential therapeutic target against AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , FN-kappa B/metabolismo , Neuroglía/metabolismo , Timosina/genética , Timosina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Memoria , Ratones , Ratones Transgénicos , Microglía/metabolismo , Neuronas/metabolismo , Fenotipo , Presenilina-1/genética , Transducción de Señal
2.
Neurobiol Learn Mem ; 168: 107154, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31904546

RESUMEN

Cognitive impairment in Alzheimer's disease (AD) is characterized by being deficient at learning and memory. Aß1-42 oligomers have been shown to impair rodent cognitive function. We previously demonstrated that activation of α7nAChR, inhibition of p38 or JNK could alleviate Aß-induced memory deficits in Y maze test. In this study, we investigated whether the effects of α7nAChR and MAPKs on Y maze test is reproducible with a hippocampus-dependent spatial memory test such as Morris water maze. We also assessed the possible co-existence of hippocampus-independent recognition memory dysfunction using a novel object recognition test and an alternative and stress free hippocampus-dependent recognition memory test such as the novel place recognition. Besides, previous research from our lab has shown that MAPKs pathways regulate Aß internalization through mediating α7nAChR. In our study, whether MAPKs pathways exert their functions in cognition by modulating α7nAChR through regulating glutamate receptors and synaptic protein, remain little known. Our results showed that activation of α7nAChR restored spatial memory, novel place recognition memory, and short-term and long-term memory in novel object recognition. Inhibition of p38 restored spatial memory and short-term and long-term memory in novel object recognition. Inhibition of ERK restored short-term memory in novel object recognition and novel place recognition memory. Inhibition of JNK restored spatial memory, short-term memory in novel object recognition and novel place recognition memory. Beside this, the activation of α7nAChR, inhibition of p38 or JNK restored Aß-induced levels of NMDAR1, NMDAR2A, NMDAR2B, GluR1, GluR2 and PSD95 in Aß-injected mice without influencing synapsin 1. In addition, these treatments also recovered the expression of acetylcholinesterase (AChE). Finally, we found that the inhibition of p38 or JNK resulted in the upregulation of α7nAChR mRNA levels in the hippocampus. Our results indicated that inhibition of p38 or JNK MAPKs could alleviate Aß-induced spatial memory deficits through regulating activation of α7nAChR via recovering memory-related proteins. Moreover, p38, ERK and JNK MAPKs exert different functions in spatial and recognition memory.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Cognición/fisiología , Sistema de Señalización de MAP Quinasas , Aprendizaje por Laberinto/fisiología , Fragmentos de Péptidos/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/administración & dosificación , Animales , Modelos Animales de Enfermedad , MAP Quinasa Quinasa 4/metabolismo , Ratones Endogámicos C57BL , Fragmentos de Péptidos/administración & dosificación , Reconocimiento en Psicología/fisiología
3.
Int Immunopharmacol ; 108: 108718, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35367744

RESUMEN

BACKGROUND: Perilipin 5 (Plin5) acts as a pivotal mediator of oxidative stress and inflammation and is associated with the progression of relevant diseases. Cerebral ischemic stroke is a severe pathological condition that involves excess oxidative stress and inflammation. However, whether Plin5 plays a role in the progression of cerebral ischemic stroke remains unaddressed. This work focused on the investigation of Plin5 in oxygen-glucose deprivation/reoxygenation (OGD/R)-injured neurons, an in vitro model for studying cerebral ischemic stroke. METHODS: The primary neuronal cells were isolated from the hippocampus of newborn mice. Neurons were subjected to OGD/R treatment to establish an in vitro model for studying cerebral ischemic stroke. Neurons were infected with recombinant adenovirus expressing Plin5 to upregulate Plin5 expression. The mRNA levels were measured by real-time quantitative PCR (RT-qPCR). Protein levels were determined by immunoblotting. Cell viability was assessed via cell counting kit-8 (CCK-8) assay. Cell apoptosis was evaluated via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and Annexin V-Allophycocyanin/7-Amino Actinomycin D (Annexin V-APC/7-AAD) apoptotic assays. Oxidative stress was monitored by dichlorofluorescein diacetate (DCFH-DA) probe. Inflammatory cytokine release was detected by enzyme-linked immunosorbent assay (ELISA). RESULTS: A decreased level of Plin5 was observed in neurons challenged with OGD/R. Plin5 overexpression remarkably subdued OGD/R-elicited apoptosis, oxidative stress and proinflammatory response. Plin5 overexpression led to an enhancement of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway associated with regulation of the Akt-glycogen synthase kinase-3ß (GSK-3ß). The blocking of Akt was able to reverse the enhancing effect of Plin5 on Nrf2 activation. The restraining of Akt or silencing of Nrf2 diminished the protective effects of Plin5 in OGD/R-injured neurons. CONCLUSIONS: Plin5 confers neuroprotection for neurons against OGD/R damage via effects on the Nrf2-Akt-GSK-3ß pathway. This work indicates a possible role of Plin5 in cerebral ischemic stroke and the up-regulation of Plin5 is a sort of survival strategy for neurons suffering from ischemic injury.


Asunto(s)
Accidente Cerebrovascular Isquémico , Perilipina-5 , Daño por Reperfusión , Animales , Anexina A5/metabolismo , Anexina A5/farmacología , Apoptosis , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamación/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas , Estrés Oxidativo , Oxígeno/metabolismo , Perilipina-5/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal
4.
Exp Neurol ; 347: 113918, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34748756

RESUMEN

In temporal lobe epilepsy (TLE), abnormal axon guidance and synapse formation lead to sprouting of mossy fibers in the hippocampus, which is one of the most consistent pathological findings in patients and animal models with TLE. Glypican 4 (Gpc4) belongs to the heparan sulfate proteoglycan family, which play an important role in axon guidance and excitatory synapse formation. However, the role of Gpc4 in the development of mossy fibers sprouting (MFS) and its underlying mechanism remain unknown. Using a pilocarpine-induced mice model of epilepsy, we showed that Gpc4 expression was significantly increased in the stratum granulosum of the dentate gyrus at 1 week after status epilepticus (SE). Using Gpc4 overexpression or Gpc4 shRNA lentivirus to regulate the Gpc4 level in the dentate gyrus, increased or decreased levels of netrin-1, SynI, PSD-95, and Timm score were observed in the dentate gyrus, indicating a crucial role of Gpc4 in modulating the development of functional MFS. The observed effects of Gpc4 on MFS were significantly antagonized when mice were treated with L-leucine or rapamycin, an agonist or antagonist of the mammalian target of rapamycin (mTOR) signal, respectively, demonstrating that mTOR pathway is an essential requirement for Gpc4-regulated MFS. Additionally, the attenuated spontaneous recurrent seizures (SRSs) were observed during chronic stage of the disease by suppressing the Gpc4 expression after SE. Altogether, our findings demonstrate a novel control of neuronal Gpc4 on the development of MFS through the mTOR pathway after pilocarpine-induced SE. Our results also strongly suggest that Gpc4 may serve as a promising target for antiepileptic studies.


Asunto(s)
Glipicanos/biosíntesis , Fibras Musgosas del Hipocampo/metabolismo , Pilocarpina/toxicidad , Transducción de Señal/fisiología , Estado Epiléptico/metabolismo , Serina-Treonina Quinasas TOR/biosíntesis , Animales , Células Cultivadas , Glipicanos/antagonistas & inhibidores , Masculino , Ratones , Fibras Musgosas del Hipocampo/efectos de los fármacos , Agonistas Muscarínicos/toxicidad , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/inducido químicamente , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
5.
Neuropeptides ; 73: 96-106, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30579679

RESUMEN

Alzheimer's disease (AD) is one of the major disabling and lethal diseases for aged individuals worldwide. To date, there are more than 10 hypotheses proposed for AD pathology. The beta-amyloid (Aß) cascade hypothesis is the most widely accepted and proposes that the accumulation of Aß in the brain is one potential mechanism for AD pathogenesis. Because some Aß-overloaded patients do not have AD syndrome, this hypothesis is challenged from time to time. More recently, it has been shown that intracellular Aß plays a key role in AD pathology. Aß is internalized by receptors distributed on the cell membrane. Among these receptors, the alpha7 nicotinic acetylcholine receptor (α7 nAChR) has been shown to play an important role in AD. The α7 nAChR is a ligand-gated ion channel and is expressed in pivotal brain regions (e.g., the cerebral cortex and hippocampus) responsible for cognitive functions. The α7 nAChR is localized both presynaptically and postsynaptically, where it activates intracellular signaling cascades. Its agonist has been investigated in clinical studies to improve cognitive functions in AD. Although many studies have shown the importance of the α7 nAChR in AD, little is known regarding its role in AD pathology. Therefore, in the current review, we summarized the basic information regarding the structures and functions of the α7 nAChR, the distribution and expression of the α7 nAChR, and the role of the α7 nAChR in mediating Aß internalization. We subsequently focused on introducing the comprehensive α7 nAChR related signaling pathways and how these signaling pathways are integrated with the α7 nAChR to play a role in AD. Finally, we stressed the AD therapy that targets the α7 nAChR.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Humanos , Transducción de Señal/fisiología
6.
Neurochem Int ; 120: 238-250, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30217465

RESUMEN

Amyloid ß peptide 1-42 (Aß1-42) could induce cognitive deficits through oxidative stress, inflammation, and neuron death in Alzheimer's disease (AD). MAPK pathways have been thought to mediate Aß1-42-induced neuroinflammation responses, neuron death and cognitive decline in AD. The α7 nicotinic acetylcholine receptor (α7nAChR) exerts a neuroprotective effect. However, whether α7nAChR alleviates Aß1-42-induced neurotoxicity through MAPKs (p38, ERK, JNK) in vivo remains unclear. In our study, memory was assessed in C57BL/6 mice using a Y-maze test. Cell death was assessed by Nissl and Hoechst staining and Bax, Bcl-2, Caspase 3, and Cytochrome C levels using Western blotting. Oxidative stress was assayed by superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA) levels. Inflammation was examined with GFAP and Iba1 using immunohistochemistry. The Aß degrading enzymes insulin degrading enzyme (IDE) and neprilysin (NEP) were tested using Western blotting. We found that activating α7nAChR or inhibiting p38 or JNK pathway alleviated Aß1-42-induced cognitive deficits and neuron loss and death by reducing oxidative stress. In addition, activating α7nAChR or inhibiting p38 or JNK pathway also reduced inflammation, which was observed as reduced GFAP and Iba1 levels with different effects on Aß degrading enzymes. Finally, we found that the activation of α7nAChR led to the downregulation of pp38 and pJNK levels. Conversely, the inhibition of p38 or JNK resulted in the upregulation of α7nAChR levels in the hippocampus and cortex. Our data indicate that the activation of α7nAChR alleviates Aß1-42-induced neurotoxicity, and this protective effect might act through the downregulation of p38 and JNK MAPKs.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Brain Res Bull ; 137: 41-52, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29128415

RESUMEN

Alzheimer's disease (AD) is one of the most devastating neurodegenerative disorders. Intracellular ß-amyloid protein (Aß) is an early event in AD. It induces the formation of amyloid plaques and neuron damage. The α7 nicotinic acetylcholine receptor (α7nAChR) has been suggested to play an important role in Aß caused cognition. It has high affinity with Aß and could mediate Aß internalization in vitro. However, whether in mouse brain the p38 MAPK signaling pathway is involved in the regulation of the α7nAChR mediated Aß internalization and their role in mitochondria remains little known. Therefore, in this study, we revealed that Aß is internalized by cholinergic and GABAergic neurons. The internalized Aß were found deposits in lysosomes/endosomes and mitochondria. Aß could form Aß-α7nAChR complex with α7nAChR, activates the p38 mitogen activated protein kinase (MAPK). And the increasing of α7nAChR could in return mediate Aß internalization in the cortex and hippocampus. In addition, by using the α7nAChR agonist PNU282987, the p38 phosphorylation level decreases, rescues the biochemical changes which are tightly associated with Aß-induced apoptosis, such as Bcl2/Bax level, cytochrome c (Cyt c) release. Collectively, the p38 MAPK signaling pathway could regulate the α7nAChR-mediated internalization of Aß. The activation of α7nAChR or the inhibition of p38 MAPK signaling pathway may be a beneficial therapy to AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Benzamidas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Compuestos Bicíclicos con Puentes/farmacología , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/patología , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/patología , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Agonistas Nicotínicos/farmacología , Fosforilación , Distribución Aleatoria , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores
8.
Brain Res ; 1663: 151-160, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28274609

RESUMEN

ß-Amyloid (Aß) accumulation in the brain is the major pathophysiology of Alzheimer disease (AD). Hypertension is a risk factor for AD by promoting Aß deposition. Traditional Chinese medicinal compound tongxinluo (TXL) can improve blood circulation and endothelium-dependent vasodilation. This study investigates the effects of TXL on cognition and Aß using spontaneously hypertensive rats (SHRs). TXL was intragastrically administered to SHRs at low-dose, mid-dose and high-dose for 15, 30 or 60days. Cognition was evaluated with a Morris Water Maze (MWM). Aß in the brain was detected by western blot, ELISA and Thioflavin-S staining. Western blot and RT-PCR were employed to exam the expression of receptor for advanced glycation end products (RAGE), low-density lipoprotein receptor-related protein-1 (LRP-1) and amyloid precursor protein (APP). After TXL treatment for 60days, compared with the vehicle, the number of crossed platform and the time spent in the target quadrant increased in parallel with the increasing length of treatment in MWM. Moreover, the Aß in the hippocampus significantly decreased compared to the vehicle group, both in western blot and ELISA. Additionally, TXL reduced RAGE expression in a dose- and time-depended manner, but LRP-1 expression had no difference between TXL groups and vehicle groups. Furthermore, the ß-secretase expression was significantly decreased compared to the vehicle group, but APP expression had no difference. In conclusion, TXL improved cognition and decreased Aß in SHRs in a dose- and time-dependent manner, the underlying mechanism may involved in inhibiting RAGE and ß-secretase expression.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Cognición/efectos de los fármacos , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/uso terapéutico , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Medicamentos Herbarios Chinos/farmacología , Hipocampo/metabolismo , Hipertensión/etiología , Hipertensión/terapia , Masculino , Medicina Tradicional China , Ratas , Ratas Endogámicas SHR , Ratas Wistar
9.
Int J Biochem Cell Biol ; 76: 75-86, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27163530

RESUMEN

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Recently, increasing evidence suggests that intracellular ß-amyloid protein (Aß) alone plays a pivotal role in the progression of AD. Therefore, understanding the signaling pathway and proteins that control Aß internalization may provide new insight for regulating Aß levels. In the present study, the regulation of Aß internalization by p38 mitogen-activated protein kinases (MAPK) through low-density lipoprotein receptor-related protein 1 (LRP1) was analyzed in vivo. The data derived from this investigation revealed that Aß1-42 were internalized by neurons and astrocytes in mouse brain, and were largely deposited in mitochondria and lysosomes, with some also being found in the endoplasmic reticulum. Aß1-42-LRP1 complex was formed during Aß1-42 internalization, and the p38 MAPK signaling pathway was activated by Aß1-42 via LRP1. Aß1-42 and LRP1 were co- localized in the cells of parietal cortex and hippocampus. Furthermore, the level of LRP1-mRNA and LRP1 protein involved in Aß1-42 internalization in mouse brain. The results of this investigation demonstrated that Aß1-42 induced an LRP1-dependent pathway that related to the activation of p38 MAPK resulting in internalization of Aß1-42. These results provide evidence supporting a key role for the p38 MAPK signaling pathway which is involved in the regulation of Aß1-42 internalization in the parietal cortex and hippocampus of mouse through LRP1 in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Hipocampo/metabolismo , Sistema de Señalización de MAP Quinasas , Neuronas/metabolismo , Lóbulo Parietal/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Femenino , Hipocampo/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Lóbulo Parietal/patología , Fragmentos de Péptidos/genética , ARN Mensajero/metabolismo , Receptores de LDL/genética , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
10.
Neurotoxicology ; 51: 166-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26511841

RESUMEN

Puerarin (PUE), an isoflavone purified from the root of Pueraria lobata (Chinese herb), has been reported to attenuate learning and memory impairments in the transgenic mouse model of Alzheimer's disease (AD). In the present study, we tested PUE in a sporadic AD (SAD) mouse model which was induced by the intracerebroventricular injection of streptozotocin (STZ). The mice were administrated PUE (25, 50, or 100mg/kg/d) for 28 days. Learning and memory abilities were assessed by the Morris water maze test. After behavioral test, the biochemical parameters of oxidative stress (glutathione peroxidase (GSH-Px), superoxide dismutases (SOD), and malondialdehyde (MDA)) were measured in the cerebral cortex and hippocampus. The SAD mice exhibited significantly decreased learning and memory ability, while PUE attenuated these impairments. The activities of GSH-Px and SOD were decreased while MDA was increased in the SAD animals. After PUE treatment, the activities of GSH-Px and SOD were elevated, and the level of MDA was decreased. The middle dose PUE was more effective than others. These results indicate that PUE attenuates learning and memory impairments and inhibits oxidative stress in STZ-induced SAD mice. PUE may be a promising therapeutic agent for SAD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Isoflavonas/administración & dosificación , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Alzheimer/inducido químicamente , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Femenino , Glutatión Peroxidasa/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Malondialdehído/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Estreptozocina , Superóxido Dismutasa/metabolismo
11.
Int J Biochem Cell Biol ; 64: 252-64, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25936756

RESUMEN

Mounting evidence suggests that the pathological hallmarks of Alzheimer's disease (AD) are caused by the intraneuronal accumulation of beta-amyloid protein (Aß). Reuptake of extracellular Aß is believed to contribute significantly to the intraneuronal Aß pool in the early stages of AD. Published reports have claimed that the low-density lipoprotein receptor-related protein 1 (LRP1) mediates Aß1-42 uptake and lysosomal trafficking in GT1-7 neuronal cells and mouse embryonic fibroblast non-neuronal cells. However, there is no direct evidence supporting the role of LRP1 in Aß internalization in primary neurons. Our recent study indicated that p38 MAPK and ERK1/2 signaling pathways are involved in regulating α7 nicotinic acetylcholine receptor (α7nAChR)-mediated Aß1-42 uptake in SH-SY5Y cells. This study was designed to explore the regulation of MAPK signaling pathways on LRP1-mediated Aß internalization in neurons. We found that extracellular Aß1-42 oligomers could be internalized into endosomes/lysosomes and mitochondria in cortical neurons. Aß1-42 and LRP1 were also found co-localized in neurons during Aß1-42 internalization, and they could form Aß1-42-LRP1 complex. Knockdown of LRP1 expression significantly decreased neuronal Aß1-42 internalization. Finally, we identified that p38 MAPK and ERK1/2 signaling pathways regulated the internalization of Aß1-42 via LRP1. Therefore, these results demonstrated that LRP1, p38 MAPK and ERK1/2 mediated the internalization of Aß1-42 in neurons and provided evidence that blockade of LRP1 or inhibitions of MAPK signaling pathways might be a potential approach to lowering brain Aß levels and served a potential therapeutic target for AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Sistema de Señalización de MAP Quinasas , Fragmentos de Péptidos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/citología , Endosomas/metabolismo , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Lisosomas/metabolismo , Ratones , Mitocondrias/metabolismo , Neuronas , Cultivo Primario de Células , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA