Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Bioorg Med Chem ; 21(13): 3821-30, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23643900

RESUMEN

In an effort to design inhibitors of human glutaminyl cyclase (QC), we have synthesized a library of N-aryl N-(5-methyl-1H-imidazol-1-yl)propyl thioureas and investigated the contribution of the aryl region of these compounds to their structure-activity relationships as cyclase inhibitors. Our design was guided by the proposed binding mode of the preferred substrate for the cyclase. In this series, compound 52 was identified as the most potent QC inhibitor with an IC50 value of 58 nM, which was two-fold more potent than the previously reported lead 2. Compound 52 is a most promising candidate for future evaluation to monitor its ability to reduce the formation of pGlu-Aß and Aß plaques in cells and transgenic animals.


Asunto(s)
Aminoaciltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Aminoaciltransferasas/metabolismo , Diseño de Fármacos , Células HEK293 , Humanos , Concentración 50 Inhibidora
2.
Biochem Biophys Res Commun ; 391(1): 756-61, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19944066

RESUMEN

Hyperglycemia induces activation of glutathione peroxidase 1 (GPX1), an anti-oxidant enzyme essential for cell survival during oxidative stress. However, the mechanism of GPX1 activation is unclear. Here, we report that hyperglycemia-induced protein glycosylation by O-linked N-acetylglucosamine (O-GlcNAc) is crucial for activation of GPX1 and for its binding to c-Abl and Arg kinases. GPX1 itself is modified with O-GlcNAc on its C-terminus. We also demonstrate that pharmacological injection of the O-GlcNAcase inhibitor NTZ induces GPX1 activation in the mouse liver. Our findings suggest a crucial role for GPX1 and its O-GlcNAc modification in hyperglycemia and diabetes mellitus.


Asunto(s)
Acetilglucosamina/metabolismo , Diabetes Mellitus/enzimología , Glutatión Peroxidasa/metabolismo , Hiperglucemia/enzimología , Acilación , Animales , Línea Celular , Activación Enzimática , Femenino , Glutatión Peroxidasa/genética , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratas , Glutatión Peroxidasa GPX1
3.
J Nanosci Nanotechnol ; 20(8): 4699-4703, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32126643

RESUMEN

In this work, noise mechanism of a tunneling field-effect transistor (TFET) on a silicon-on-insulator substrate was studied as a function of temperature. The results show that the drain current and subthreshold slope increase with increase in temperature. This temperature dependence is likely caused by the generation of greater current flow owing to decreased silicon band gap and leakage. Further, the TFET noise decreases with increase in temperature. Therefore, the effective tunneling length between the source and the channel appears to decrease and Poole-Frenkel tunneling occurs.

4.
Aging Cell ; 18(1): e12872, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30515991

RESUMEN

Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss resulting from cumulative neuronal cell death. O-linked ß-N-acetyl glucosamine (O-GlcNAc) modification of the proteins reflecting glucose metabolism is altered in the brains of patients with AD. However, the link between altered O-GlcNAc modification and neuronal cell death in AD is poorly understood. Here, we examined the regulation of O-GlcNAcylation of c-Fos and the effects of O-GlcNAcylated c-Fos on neuronal cell death during AD pathogenesis. We found that amyloid beta (Aß)-induced O-GlcNAcylation on serine-56 and 57 of c-Fos was resulted from decreased interaction between c-Fos and O-GlcNAcase and promoted neuronal cell death. O-GlcNAcylated c-Fos increased its stability and potentiated the transcriptional activity through higher interaction with c-Jun, resulting in induction of Bim expression leading to neuronal cell death. Taken together, Aß-induced O-GlcNAcylation of c-Fos plays an important role in neuronal cell death during the pathogenesis of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/toxicidad , Animales , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glicosilación/efectos de los fármacos , Humanos , Ratones Transgénicos , Neuronas/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos , beta-N-Acetilhexosaminidasas/metabolismo
5.
Biochem Biophys Res Commun ; 372(4): 913-7, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18538664

RESUMEN

Beta amyloid peptide is generated from amyloid precursor protein (APP) by proteolytic cleavage of beta- and gamma-secretases, and plays a critical role in the pathogenesis of Alzheimer's disease. Since gamma-secretase cleaves several proteins including APP and Notch in a number of cell types, it is important to understand the conditions determining gamma-secretase substrate specificity. In the present study, inhibition of Rac1 attenuated gamma-secretase activity for APP, resulting in decreased production of the APP intracellular domain but accumulated C-terminal fragments (APP-CTF). In contrast, Rac1 inhibitor, NSC23766 increased production of the Notch1 intracellular domain but slightly decreased the ectodomain-shed form of Notch1 (NotchDeltaE). To elucidate the mechanism underlying these observations, we performed co-immunoprecipitation experiments to analyze the interaction between Rac1 and presenilin1 (PS1), a component of the gamma-secretase complex. Inhibition of Rac1 enhanced its interaction with PS1. Under the same condition, PS1 interacted more strongly with NotchDeltaE than with APP-CTF. Our results suggested that PS1 determines the preferred substrate for gamma-secretase between APP and Notch1, depending on the activation status of Rac1.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Receptor Notch1/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Aminoquinolinas/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Diseño de Fármacos , Activación Enzimática , Humanos , Presenilina-1/metabolismo , Estructura Terciaria de Proteína , Pirimidinas/farmacología , Receptor Notch1/genética , Eliminación de Secuencia , Especificidad por Sustrato , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
6.
Mech Ageing Dev ; 161(Pt A): 83-94, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27105872

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. Autosomal dominant, familial AD (fAD) is very rare and caused by mutations in amyloid precursor protein (APP), presenilin-1 (PSEN-1), and presenilin-2 (PSEN-2) genes. The pathogenesis of sporadic AD (sAD) is more complex and variants of several genes are associated with an increased lifetime risk of AD. Nuclear and mitochondrial DNA integrity is pivotal during neuronal development, maintenance and function. DNA damage and alterations in cellular DNA repair capacity have been implicated in the aging process and in age-associated neurodegenerative diseases, including AD. These findings are supported by research using animal models of AD and in DNA repair deficient animal models. In recent years, novel mechanisms linking DNA damage to neuronal dysfunction have been identified and have led to the development of noninvasive treatment strategies. Further investigations into the molecular mechanisms connecting DNA damage to AD pathology may help to develop novel treatment strategies for this debilitating disease. Here we provide an overview of the role of genome instability and DNA repair deficiency in AD pathology and discuss research strategies that include genome instability as a component.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Inestabilidad Genómica , Mutación , Neuronas/metabolismo , Presenilina-1/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Daño del ADN , Reparación del ADN , Humanos , Neuronas/patología , Presenilina-1/genética
7.
Stem Cells Transl Med ; 6(1): 293-305, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28170178

RESUMEN

Transplantation of stem cells into the brain attenuates functional deficits in the central nervous system via cell replacement, the release of specific neurotransmitters, and the production of neurotrophic factors. To identify patient-specific and safe stem cells for treating Alzheimer's disease (AD), we generated induced pluripotent stem cells (iPSCs) derived from mouse skin fibroblasts by treating protein extracts of embryonic stem cells. These reprogrammed cells were pluripotent but nontumorigenic. Here, we report that protein-iPSCs differentiated into glial cells and decreased plaque depositions in the 5XFAD transgenic AD mouse model. We also found that transplanted protein-iPSCs mitigated the cognitive dysfunction observed in these mice. Proteomic analysis revealed that oligodendrocyte-related genes were upregulated in brains injected with protein-iPSCs, providing new insights into the potential function of protein-iPSCs. Taken together, our data indicate that protein-iPSCs might be a promising therapeutic approach for AD. Stem Cells Translational Medicine 2017;6:293-305.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/terapia , Células Madre Pluripotentes Inducidas/trasplante , Animales , Conducta Animal , Encéfalo/patología , Diferenciación Celular , Disfunción Cognitiva/complicaciones , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Oligodendroglía/metabolismo , Placa Amiloide/patología , Proteoma/metabolismo , Trasplante de Células Madre , Transferrina/metabolismo , Regulación hacia Arriba/genética
8.
Eur J Med Chem ; 125: 1172-1192, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27855359

RESUMEN

In this study, we designed a library of compounds based on the structures of well-known ligands of the 18 kDa translocator protein (TSPO), one of the putative components of the mPTP. We performed diverse mitochondrial functional assays to assess their ability to restore cells from Aß-induced toxicity in vitro and in vivo. Among tested compounds, compound 25 effectively improved cognitive function in animal models of AD. Given the excellent in vitro and in vivo activity and a favorable pharmacokinetic profile of compound 25, we believe that it can serve as a promising lead compound for a potential treatment option for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Bencimidazoles/química , Bencimidazoles/uso terapéutico , Mitocondrias/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Bencimidazoles/farmacocinética , Bencimidazoles/farmacología , Cognición/efectos de los fármacos , Humanos , Ligandos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Modelos Moleculares , Ratas Sprague-Dawley , Receptores de GABA/metabolismo
9.
Sci Rep ; 6: 25628, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27156849

RESUMEN

Alzheimer's disease (AD) is the most common type of dementia and age-related neurodegenerative disease. Elucidating the cellular changes that occur during ageing is an important step towards understanding the pathogenesis and progression of neurodegenerative disorders. SIRT6 is a member of the mammalian sirtuin family of anti-aging genes. However, the relationship between SIRT6 and AD has not yet been elucidated. Here, we report that SIRT6 protein expression levels are reduced in the brains of both the 5XFAD AD mouse model and AD patients. Aß42, a major component of senile plaques, decreases SIRT6 expression, and Aß42-induced DNA damage is prevented by the overexpression of SIRT6 in HT22 mouse hippocampal neurons. Also, there is a strong negative correlation between Aß42-induced DNA damage and p53 levels, a protein involved in DNA repair and apoptosis. In addition, upregulation of p53 protein by Nutlin-3 prevents SIRT6 reduction and DNA damage induced by Aß42. Taken together, this study reveals that p53-dependent SIRT6 expression protects cells from Aß42-induced DNA damage, making SIRT6 a promising new therapeutic target for the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Daño del ADN , Sirtuinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Antracenos/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Células HCT116 , Histonas/metabolismo , Humanos , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lisina/metabolismo , Ratones , Ratones Transgénicos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley
10.
Metabolism ; 65(4): 533-42, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26975545

RESUMEN

OBJECTIVE: We investigated the effect and regulatory mechanism of 2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone (DMC) isolated from Cleistocalyx operculatus on metabolic parameters in myotubes, adipocytes and an obese mouse model. MATERIALS AND METHODS: Myotubes and adipocytes were incubated with or without DMC. Glucose uptake, fatty acid oxidation, AMPK activation and adipocytes differentiation were investigated. To examine in vivo effect of DMC, 30mg/kg/day DMC was administered by oral gavage for 2weeks in high fat fed C57BL/6 male mice and intra-peritoneal glucose tolerance test was performed. In order to examine whether DMC directly activates AMPK, we performed cell free AMPK assay and surface plasmon resonance spectroscopy analysis. RESULT: DMC increases glucose uptake and fatty acid oxidation (FAO) in myotubes. Also, DMC inhibits adipocyte differentiation in 3T3-L1 cells. Interestingly, DMC stimulates phosphorylation of AMP-dependent protein kinase (AMPK) alpha subunit (T172) by directly binding to AMPK, which results in the activation of AMPK. Furthermore, DMC binds AMPK with a higher affinity than AMP. When AMPK was knocked down, the stimulatory effect of DMC on FAO and its inhibitory effect on adipogenesis were abolished. These results suggest that the effects of DMC were primarily mediated by AMPK activation. In addition, treating mice fed a high fat diet with DMC improved glucose tolerance and significantly increased FAO of the muscles. CONCLUSION: DMC, as a novel AMPK activator, shows anti-diabetic effects in cell culture systems, such as myotubes and adipocytes, and in a diet-induced obese mouse model.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Chalconas/uso terapéutico , Activadores de Enzimas/uso terapéutico , Intolerancia a la Glucosa/tratamiento farmacológico , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Animales , Diferenciación Celular , Células Cultivadas , Chalconas/aislamiento & purificación , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Obesidad/metabolismo , Syzygium/química
11.
Mol Neurodegener ; 11: 4, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26758977

RESUMEN

BACKGROUND: The receptor for advanced glycation end products (RAGE) has been found to interact with amyloid ß (Aß). Although RAGE does not have any kinase motifs in its cytosolic domain, the interaction between RAGE and Aß triggers multiple cellular signaling involved in Alzheimer's disease (AD). However, the mechanism of signal transduction by RAGE remains still unknown. Therefore, identifying binding proteins of RAGE may provide novel therapeutic targets for AD. RESULTS: In this study, we identified p38-regulated/activated protein kinase (PRAK) as a novel RAGE interacting molecule. To investigate the effect of Aß on PRAK mediated RAGE signaling pathway, we treated SH-SY5Y cells with monomeric form of Aß. We demonstrated that Aß significantly increased the phosphorylation of PRAK as well as the interaction between PRAK and RAGE. We showed that knockdown of PRAK rescued mTORC1 inactivation induced by Aß treatment and decreased the formation of Aß-induced autophagosome. CONCLUSIONS: We provide evidence that PRAK plays a critical role in AD pathology as a key interactor of RAGE. Thus, our data suggest that PRAK might be a potential therapeutic target of AD involved in RAGE-mediated cell signaling induced by Aß.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Autofagia/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal , Péptidos beta-Amiloides/metabolismo , Humanos , Neuronas/metabolismo , Fosforilación , Transducción de Señal/fisiología
12.
Mol Neurodegener ; 10: 13, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25888034

RESUMEN

BACKGROUND: Patients with Alzheimer's disease (AD) frequently experience disruption of their circadian rhythms, but whether and how circadian clock molecules are perturbed by AD remains unknown. AD is an age-related neurological disorder and amyloid-ß (Aß) is one of major causative molecules in the pathogenesis of AD. RESULTS: In this study, we investigated the role of Aß in the regulation of clock molecules and circadian rhythm using an AD mouse model. These mice exhibited altered circadian behavior, and altered expression patterns of the circadian clock genes, Bmal1 and Per2. Using cultured cells, we showed that Aß induces post-translational degradation of the circadian clock regulator CBP, as well as the transcription factor BMAL1, which forms a complex with the master circadian transcription factor CLOCK. Aß-induced degradation of BMAL1 and CBP correlated with the reduced binding of transcription factors to the Per2 promoter, which in turn resulted in disruptions to PER2 protein expression and the oscillation of Per2 mRNA levels. CONCLUSIONS: Our results elucidate the underlying mechanisms for disrupted circadian rhythm in AD.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Péptidos beta-Amiloides/metabolismo , Ritmo Circadiano/genética , Proteínas de la Membrana/metabolismo , Proteínas Circadianas Period/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción ARNTL/genética , Enfermedad de Alzheimer/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de la Membrana/genética , Ratones Transgénicos , Fosfoproteínas/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
13.
J Alzheimers Dis ; 43(3): 797-807, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25114069

RESUMEN

Alzheimer's disease is the most prevalent neurodegenerative disorder, characterized by neurofibrillary tangles, senile plaques, and neuron loss. Amyloid beta peptides are generated from amyloid beta precursor protein by consecutive catalysis by ß and γ-secretases. Diversely modified forms of A have been N3pE-42 Aß has received considerable attention as one of the major constituents of the senile plaques of AD brains due to its higher aggregation velocity, stability, and hydrophobicity compared to the full-length A. A previous study suggested that is catalyzed by glutaminyl cyclase (QC) following limited proteolysis of Aß at the N-terminus. Here, we reveal that decreasing the QC activity via application of a QC inhibitor modulates-γ-secretase activity, resulting in diminished plaque formation as well as reduced N3pE 42 Aß aggregates in the subiculum of the 5XFAD mouse model of AD. This study suggests a possible novel mechanism by which QC regulates Aß formation , namely modulation of γ-secretase activity.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aminoaciltransferasas/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Placa Amiloide/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología
14.
Chem Commun (Camb) ; 49(13): 1303-5, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23295364

RESUMEN

We report a small molecule two-photon probe (SAD1) that shows a significant TP action cross section (170 GM), binds to Aß plaques specifically, readily enters the brain through the BBB, and can directly 3D monitor the individual Aß plaque in living transgenic mice at more than 380 µm depths.


Asunto(s)
2-Naftilamina/análogos & derivados , Benzotiazoles/química , Benzotiazoles/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Naftalenos/química , Naftalenos/metabolismo , Fotones , Placa Amiloide/metabolismo , 2-Naftilamina/química , 2-Naftilamina/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Imagenología Tridimensional , Ratones , Peso Molecular , Placa Amiloide/patología , Solubilidad
15.
Neurobiol Aging ; 34(1): 275-85, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22503002

RESUMEN

Deposition of ß-amyloid (Aß) as senile plaques and disrupted glucose metabolism are two main characteristics of Alzheimer's disease (AD). It is unknown, however, how these two processes are related in AD. Here we examined the relationship between O-GlcNAcylation, which is a glucose level-dependent post-translational modification that adds O-linked ß-N-acetylglucosamine (O-GlcNAc) to proteins, and Aß production in a mouse model of AD carrying 5XFAD genes. We found that 1,2-dideoxy-2'-propyl-α-d-glucopyranoso-[2,1-D]-Δ2'-thiazoline (NButGT), a specific inhibitor of O-GlcNAcase, reduces Aß production by lowering γ-secretase activity both in vitro and in vivo. We also found that O-GlcNAcylation takes place at the S708 residue of nicastrin, which is a component of γ-secretase. Moreover, NButGT attenuated the accumulation of Aß, neuroinflammation, and memory impairment in the 5XFAD mice. This is the first study to show the relationship between Aß generation and O-GlcNAcylation in vivo. These results suggest that O-GlcNAcylation may be a suitable therapeutic target for the treatment of AD.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/enzimología , beta-N-Acetilhexosaminidasas/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Condicionamiento Psicológico/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Miedo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Ratones , Ratones Transgénicos , Mutación/genética , Fragmentos de Péptidos/metabolismo , Placa Amiloide/tratamiento farmacológico , Presenilina-1/genética , Presenilina-2/genética , Transfección
16.
Autophagy ; 8(12): 1842-4, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22931791

RESUMEN

Alzheimer disease (AD) is sometimes referred to as type III diabetes because of the shared risk factors for the two disorders. Insulin resistance, one of the major components of type II diabetes mellitus (T2DM), is a known risk factor for AD. Insulin resistance increases amyloid-ß peptide (Aß) generation, but the exact mechanism underlying the linkage of insulin resistance to increased Aß generation in the brain is unknown. In this study, we investigated the effect of insulin resistance on amyloid ß (A4) precursor protein (APP) processing in mice fed a high-fat diet (HFD), and diabetic db/db mice. We found that insulin resistance promotes Aß generation in the brain via altered insulin signal transduction, increased BACE1/ß-secretase and γ-secretase activities, and accumulation of autophagosomes. Using an in vitro model of insulin resistance, we found that defects in insulin signal transduction affect autophagic flux by inhibiting the mechanistic target of rapamycin (MTOR) pathway. The insulin resistance-induced autophagosome accumulation resulted in alteration of APP processing through enrichment of secretase proteins in autophagosomes. We speculate that the insulin resistance that underlies the pathogenesis of T2DM might alter APP processing through autophagy activation, which might be involved in the pathogenesis of AD. Therefore, we propose that insulin resistance-induced autophagosome accumulation becomes a potential linker between AD and T2DM.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Autofagia , Resistencia a la Insulina , Fagosomas/metabolismo , Procesamiento Proteico-Postraduccional , Enfermedad de Alzheimer/patología , Animales , Diabetes Mellitus Tipo 2/patología , Humanos , Ratones , Modelos Biológicos
17.
Diabetes ; 61(12): 3126-38, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22829447

RESUMEN

Insulin resistance, one of the major components of type 2 diabetes mellitus (T2DM), is a known risk factor for Alzheimer's disease (AD), which is characterized by an abnormal accumulation of intra- and extracellular amyloid ß peptide (Aß). Insulin resistance is known to increase Aß generation, but the underlying mechanism that links insulin resistance to increased Aß generation is unknown. In this study, we examined the effect of high-fat diet-induced insulin resistance on amyloid precursor protein (APP) processing in mouse brains. We found that the induced insulin resistance promoted Aß generation in the brain via altered insulin signal transduction, increased ß- and γ-secretase activities, and accumulation of autophagosomes. These findings were confirmed in diabetic db/db mice brains. Furthermore, in vitro experiments in insulin-resistant SH-SY5Y cells and primary cortical neurons confirmed the alteration of APP processing by insulin resistance-induced autophagosome accumulation. Defects in insulin signal transduction affect autophagic flux by inhibiting the mammalian target of rapamycin pathway, resulting in altered APP processing in these cell culture systems. Thus, the insulin resistance that underlies the pathogenesis of T2DM might also trigger accumulation of autophagosomes, leading to increased Aß generation, which might be involved in the pathogenesis of AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Fagosomas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Inmunohistoquímica , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética
18.
J Alzheimers Dis ; 29(3): 615-28, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22269161

RESUMEN

One of the major hallmarks of Alzheimer's disease (AD) is the extracellular deposition of amyloid-ß (Aß) as senile plaques in specific brain regions. Clearly, an understanding of the cellular processes underlying Aß deposition is a crucial issue in the field of AD research. Recent studies have found that accumulation of intraneuronal Aß (iAß) is associated with synaptic deficits, neuronal death, and cognitive dysfunction in AD patients. In this study, we found that Aß deposits had several shapes and sizes, and that iAß occurred before the formation of extracellular amyloid plaques in the subiculum of 5XFAD mice, an animal model of AD. We also observed pyroglutamate-modified Aß (N3pE-Aß), which has been suggested to be a seeding molecule for senile plaques, inside the Aß plaques only after iAß accumulation, which argues against its seeding role. In addition, we found that iAß accumulates in calcium-binding protein (CBP)-free neurons, induces neuronal death, and then develops into senile plaques in 2-4-month-old 5XFAD mice. These findings suggest that N3pE-Aß-independent accumulation of Aß in CBP-free neurons might be an early process that triggers neuronal damage and senile plaque formation in AD patients. Our results provide new insights into several long-standing gaps in AD research, namely how Aß plaques are formed, what happens to iAß and how Aß causes selective neuronal loss in AD patients.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Proteínas de Unión al Calcio/deficiencia , Hipocampo/patología , Líquido Intracelular/metabolismo , Neuronas/citología , Factores de Edad , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Humanos , Líquido Intracelular/efectos de los fármacos , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ácido Pirrolidona Carboxílico/farmacología
19.
PLoS One ; 4(1): e4172, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19137062

RESUMEN

Presenilin (PS) is involved in many cellular events under physiological and pathological conditions. Previous reports have revealed that PS deficiency results in hyperproliferation and resistance to apoptotic cell death. In the present study, we investigated the effects of PS on beta-catenin and cell mortality during serum deprivation. Under these conditions, PS1/PS2 double-knockout MEFs showed aberrant accumulation of phospho-beta-catenin, higher ROS generation, and notable cell death. Inhibition of beta-catenin phosphorylation by LiCl reversed ROS generation and cell death in PS deficient cells. In addition, the K19/49R mutant form of beta-catenin, which undergoes normal phosphorylation but not ubiquitination, induced cytotoxicity, while the phosphorylation deficient S37A beta-catenin mutant failed to induce cytotoxicity. These results indicate that aberrant accumulation of phospho-beta-catenin underlies ROS-mediated cell death in the absence of PS. We propose that the regulation of beta-catenin is useful for identifying therapeutic targets of hyperproliferative diseases and other degenerative conditions.


Asunto(s)
Muerte Celular/fisiología , Línea Celular/metabolismo , Presenilinas/deficiencia , Especies Reactivas de Oxígeno/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosforilación , Presenilinas/genética , Presenilinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA