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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Neurosci ; 88: 292-299, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29501719

RESUMEN

Stress during pregnancy increases the risk to develop psychological disorders such as depression during pregnancy or in the postpartum period. According to the neurotrophin hypothesis of depression, the pathophysiology of depression is caused by reduced neurotrophic activity in the brain. However, most studies only focus on the molecular changes happening to the offspring upon gestational stress. To gain insight into the potential molecular changes happening in the stressed dams, C57Bl6/J mice were stressed during their first week of gestation. At 28 days postpartum, the hippocampus and nucleus accumbens core of the dams, two brain regions heavily implicated in depression, were evaluated using immunohistochemistry to detect changes in the neurotrophin system. Gestational stress decreased the weight of the dams, increased the chance for spontaneous abortion and increased the weight of offspring. Litter size, survival rates and sex distribution were not altered as a consequence of gestational stress. Hippocampal brain-derived neurotrophic factor (BDNF) decreased following exposure to stress during pregnancy. Hippocampal protein levels of p75NTR, a low-affinity receptor for BDNF which can induce apoptosis, were increased following exposure to stress. Protein levels of p11, of which the expression is regulated by BDNF, were decreased in the hippocampus. No changes were found for TrkB immunostaining or apoptosis. Taken together, this shows that stress during pregnancy negatively affects the neurotrophin system in the hippocampus of the dams, thereby reducing hippocampal plasticity. These data confirm that gestational stress has a negative impact on pregnancy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Péptidos de Penetración Celular/metabolismo , Hipocampo/metabolismo , Periodo Posparto/psicología , Animales , Apoptosis/fisiología , Conducta Animal , Corticosterona/metabolismo , Femenino , Ratones Endogámicos C57BL , Embarazo , Estrés Psicológico/fisiopatología
2.
Alzheimers Dement ; 14(10): 1261-1280, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30036493

RESUMEN

INTRODUCTION: Tauopathies are neurodegenerative diseases characterized by TAU protein-related pathology, including frontotemporal dementia and Alzheimer's disease among others. Mutant TAU animal models are available, but none of them faithfully recapitulates human pathology and are not suitable for drug screening. METHODS: To create a new in vitro tauopathy model, we generated a footprint-free triple MAPT-mutant human induced pluripotent stem cell line (N279K, P301L, and E10+16 mutations) using clustered regularly interspaced short palindromic repeats-FokI and piggyBac transposase technology. RESULTS: Mutant neurons expressed pathogenic 4R and phosphorylated TAU, endogenously triggered TAU aggregation, and had increased electrophysiological activity. TAU-mutant cells presented deficiencies in neurite outgrowth, aberrant sequence of differentiation to cortical neurons, and a significant activation of stress response pathways. RNA sequencing confirmed stress activation, demonstrated a shift toward GABAergic identity, and an upregulation of neurodegenerative pathways. DISCUSSION: In summary, we generated a novel in vitro human induced pluripotent stem cell TAU-mutant model displaying neurodegenerative disease phenotypes that could be used for disease modeling and drug screening.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/patología , Potenciales de la Membrana/fisiología , Mutación , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neurogénesis/fisiología , Proyección Neuronal/fisiología , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Tauopatías/genética , Tauopatías/patología , Transcriptoma , Proteínas tau/genética
3.
Science ; 381(6663): 1176-1182, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37708272

RESUMEN

Neuronal cell loss is a defining feature of Alzheimer's disease (AD), but the underlying mechanisms remain unclear. We xenografted human or mouse neurons into the brain of a mouse model of AD. Only human neurons displayed tangles, Gallyas silver staining, granulovacuolar neurodegeneration (GVD), phosphorylated tau blood biomarkers, and considerable neuronal cell loss. The long noncoding RNA MEG3 was strongly up-regulated in human neurons. This neuron-specific long noncoding RNA is also up-regulated in AD patients. MEG3 expression alone was sufficient to induce necroptosis in human neurons in vitro. Down-regulation of MEG3 and inhibition of necroptosis using pharmacological or genetic manipulation of receptor-interacting protein kinase 1 (RIPK1), RIPK3, or mixed lineage kinase domain-like protein (MLKL) rescued neuronal cell loss in xenografted human neurons. This model suggests potential therapeutic approaches for AD and reveals a human-specific vulnerability to AD.


Asunto(s)
Enfermedad de Alzheimer , Necroptosis , Neuronas , ARN Largo no Codificante , Animales , Humanos , Ratones , Enfermedad de Alzheimer/patología , Xenoinjertos , Necroptosis/genética , Neuronas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
4.
iScience ; 26(6): 106829, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37250784

RESUMEN

microRNA-132 (miR-132), a known neuronal regulator, is one of the most robustly downregulated microRNAs (miRNAs) in the brain of Alzheimer's disease (AD) patients. Increasing miR-132 in AD mouse brain ameliorates amyloid and Tau pathologies, and also restores adult hippocampal neurogenesis and memory deficits. However, the functional pleiotropy of miRNAs requires in-depth analysis of the effects of miR-132 supplementation before it can be moved forward for AD therapy. We employ here miR-132 loss- and gain-of-function approaches using single-cell transcriptomics, proteomics, and in silico AGO-CLIP datasets to identify molecular pathways targeted by miR-132 in mouse hippocampus. We find that miR-132 modulation significantly affects the transition of microglia from a disease-associated to a homeostatic cell state. We confirm the regulatory role of miR-132 in shifting microglial cell states using human microglial cultures derived from induced pluripotent stem cells.

5.
Front Mol Neurosci ; 14: 714768, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34349622

RESUMEN

Recent advances in RNA sequencing technologies helped to uncover the existence of tens of thousands of long non-coding RNAs (lncRNAs) that arise from the dark matter of the genome. These lncRNAs were originally thought to be transcriptional noise but an increasing number of studies demonstrate that these transcripts can modulate protein-coding gene expression by a wide variety of transcriptional and post-transcriptional mechanisms. The spatiotemporal regulation of lncRNA expression is particularly evident in the central nervous system, suggesting that they may directly contribute to specific brain processes, including neurogenesis and cellular homeostasis. Not surprisingly, lncRNAs are therefore gaining attention as putative novel therapeutic targets for disorders of the brain. In this review, we summarize the recent insights into the functions of lncRNAs in the brain, their role in neuronal maintenance, and their potential contribution to disease. We conclude this review by postulating how these RNA molecules can be targeted for the treatment of yet incurable neurological disorders.

6.
Nat Commun ; 12(1): 4506, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301930

RESUMEN

Polygenic Risk Scores (PRS) for AD offer unique possibilities for reliable identification of individuals at high and low risk of AD. However, there is little agreement in the field as to what approach should be used for genetic risk score calculations, how to model the effect of APOE, what the optimal p-value threshold (pT) for SNP selection is and how to compare scores between studies and methods. We show that the best prediction accuracy is achieved with a model with two predictors (APOE and PRS excluding APOE region) with pT<0.1 for SNP selection. Prediction accuracy in a sample across different PRS approaches is similar, but individuals' scores and their associated ranking differ. We show that standardising PRS against the population mean, as opposed to the sample mean, makes the individuals' scores comparable between studies. Our work highlights the best strategies for polygenic profiling when assessing individuals for AD risk.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteínas E/genética , Estudio de Asociación del Genoma Completo/métodos , Herencia Multifactorial/genética , Polimorfismo de Nucleótido Simple , Alelos , Enfermedad de Alzheimer/diagnóstico , Estudios de Casos y Controles , Frecuencia de los Genes , Genética de Población/métodos , Genética de Población/estadística & datos numéricos , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Genotipo , Humanos , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Medición de Riesgo/estadística & datos numéricos , Factores de Riesgo , Sensibilidad y Especificidad
7.
Genes Brain Behav ; 20(1): e12695, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32812350

RESUMEN

The relative lack of sensitive and clinically valid tests of rodent behavior might be one of the reasons for the limited success of the clinical translation of preclinical Alzheimer's disease (AD) research findings. There is a general interest in innovative behavioral methodology, and protocols have been proposed for touchscreen operant chambers that might be superior to existing cognitive assessment methods. We assessed and analyzed touchscreen performance in several novel ways to examine the possible occurrence of early signs of prefrontal (PFC) functional decline in the APP/PS1 mouse model of AD. Touchscreen learning performance was compared between APP/PS1-21 mice and wildtype littermates on a C57BL/6J background at 3, 6 and 12 months of age in parallel to the assessment of spatial learning, memory and cognitive flexibility in the Morris water maze (MWM). We found that older mice generally needed more training sessions to complete the touchscreen protocol than younger ones. Older mice also displayed defects in MWM working memory performance, but touchscreen protocols detected functional changes beginning at 3 months of age. Histological changes in PFC of APP/PS1 mice indeed occurred as early as 3 months. Our results suggest that touchscreen operant protocols are more sensitive to PFC dysfunction, which is of relevance to the use of these tasks and devices in preclinical AD research and experimental pharmacology.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Investigación Conductal/métodos , Condicionamiento Operante , Aprendizaje por Laberinto , Corteza Prefrontal/fisiopatología , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Investigación Conductal/instrumentación , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Presenilina-1/genética , Interfaz Usuario-Computador
8.
Science ; 370(6512): 61-66, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33004512

RESUMEN

To provide better prevention and treatment, we need to understand the environmental and genetic risks of Alzheimer's disease (AD). However, the definition of AD has been confounded with dementia in many studies. Thus, overinterpretation of genetic findings with regard to mechanisms and drug targets may explain, in part, controversies in the field. Here, we analyze the different forms of genetic risk of AD and how these can be used to model disease. We stress the importance of studying gene variants in the right cell types and in the right pathological context. The lack of mechanistic understanding of genetic variation has become the major bottleneck in the search for new drug targets for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Desarrollo de Medicamentos , Predisposición Genética a la Enfermedad , Investigación Biomédica Traslacional , Enfermedad de Alzheimer/prevención & control , Animales , Modelos Animales de Enfermedad , Variación Genética , Humanos , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Riesgo
9.
EMBO Mol Med ; 12(3): e10606, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31951107

RESUMEN

Polygenic risk scores have identified that genetic variants without genome-wide significance still add to the genetic risk of developing Alzheimer's disease (AD). Whether and how subthreshold risk loci translate into relevant disease pathways is unknown. We investigate here the involvement of AD risk variants in the transcriptional responses of two mouse models: APPswe/PS1L166P and Thy-TAU22. A unique gene expression module, highly enriched for AD risk genes, is specifically responsive to Aß but not TAU pathology. We identify in this module 7 established AD risk genes (APOE, CLU, INPP5D, CD33, PLCG2, SPI1, and FCER1G) and 11 AD GWAS genes below the genome-wide significance threshold (GPC2, TREML2, SYK, GRN, SLC2A5, SAMSN1, PYDC1, HEXB, RRBP1, LYN, and BLNK), that become significantly upregulated when exposed to Aß. Single microglia sequencing confirms that Aß, not TAU, pathology induces marked transcriptional changes in microglia, including increased proportions of activated microglia. We conclude that genetic risk of AD functionally translates into different microglia pathway responses to Aß pathology, placing AD genetic risk downstream of the amyloid pathway but upstream of TAU pathology.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Microglía , Proteínas tau , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Hipocampo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Proteínas tau/metabolismo
10.
Nat Neurosci ; 22(12): 2111-2116, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659342

RESUMEN

Although genetics highlights the role of microglia in Alzheimer's disease, one-third of putative Alzheimer's disease risk genes lack adequate mouse orthologs. Here we successfully engraft human microglia derived from embryonic stem cells in the mouse brain. The cells recapitulate transcriptionally human primary microglia ex vivo and show expression of human-specific Alzheimer's disease risk genes. Oligomeric amyloid-ß induces a divergent response in human versus mouse microglia. This model can be used to study the role of microglia in neurological diseases.


Asunto(s)
Enfermedad de Alzheimer/genética , Células Madre Embrionarias/citología , Microglía/metabolismo , Microglía/trasplante , Transcriptoma , Péptidos beta-Amiloides/farmacología , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos
11.
Mol Neurodegener ; 13(1): 54, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30314521

RESUMEN

BACKGROUND: Despite diverging levels of amyloid-ß (Aß) and TAU pathology, different mouse models, as well as sporadic AD patients show predictable patterns of episodic memory loss. MicroRNA (miRNA) deregulation is well established in AD brain but it is unclear whether Aß or TAU pathology drives those alterations and whether miRNA changes contribute to cognitive decline. METHODS: miRNAseq was performed on cognitively intact (4 months) and impaired (10 months) male APPtg (APPswe/PS1L166P) and TAUtg (THY-Tau22) mice and their wild-type littermates (APPwt and TAUwt). We analyzed the hippocampi of 12 mice per experimental group (n = 96 in total), and employed a 2-way linear model to extract differentially expressed miRNAs. Results were confirmed by qPCR in a separate cohort of 4 M and 10 M APPtg and APPwt mice (n = 7-9 per group) and in human sporadic AD and non-demented control brain. Fluorescent in situ hybridization identified their cellular expression. Functional annotation of predicted targets was performed using GO enrichment. Behavior of wild-type mice was assessed after intracerebroventricular infusion of miRNA mimics. RESULTS: Six miRNAs (miR-10a-5p, miR-142a-5p, miR-146a-5p, miR-155-5p, miR-211-5p, miR-455-5p) are commonly upregulated between APPtg and TAUtg mice, and four of these (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) are altered in AD patients. All 6 miRNAs are strongly enriched in neurons. Upregulating these miRNAs in wild-type mice is however not causing AD-related cognitive disturbances. CONCLUSION: Diverging AD-related neuropathologies induce common disturbances in the expression of neuronal miRNAs. 4 of these miRNAs are also upregulated in AD patients. Therefore these 4 miRNAs (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) appear part of a core pathological process in AD patients and APPtg and TAUtg mice. They are however not causing cognitive disturbances in wild-type mice. As some of these miRNA target AD relevant proteins, they may be, in contrast, part of a protective response in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , MicroARNs/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Ratones Transgénicos , Neuronas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Regulación hacia Arriba
12.
EMBO Mol Med ; 8(9): 1005-18, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27485122

RESUMEN

microRNA-132 (miR-132) is involved in prosurvival, anti-inflammatory and memory-promoting functions in the nervous system and has been found consistently downregulated in Alzheimer's disease (AD). Whether and how miR-132 deficiency impacts AD pathology remains, however, unaddressed. We show here that miR-132 loss exacerbates both amyloid and TAU pathology via inositol 1,4,5-trisphosphate 3-kinase B (ITPKB) upregulation in an AD mouse model. This leads to increased ERK1/2 and BACE1 activity and elevated TAU phosphorylation. We confirm downregulation of miR-132 and upregulation of ITPKB in three distinct human AD patient cohorts, indicating the pathological relevance of this pathway in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Encéfalo/patología , MicroARNs/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas tau/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional
13.
J Alzheimers Dis ; 51(3): 677-82, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890781

RESUMEN

Treatment of Alzheimer's disease (AD) patients with the antidepressant fluoxetine is known to improve memory and cognitive function. However, the mechanisms underlying these effects are largely unknown. To unravel these mechanisms, we aimed to treat APPswe/PS1dE9 mice with fluoxetine. Unexpectedly, with time, an increased number of animals displayed seizure behavior and died. Although spontaneous behavioral seizures have been reported previously in this mouse model, the observation of seizures and death consequential to fluoxetine treatment is new. Our results warrant further research on the underlying mechanisms as this may refine the treatment of AD patients.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Fluoxetina/farmacología , Fármacos Neuroprotectores/farmacología , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticonvulsivantes/farmacología , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Modelos Logísticos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética , Presenilina-1/metabolismo
14.
PLoS One ; 9(10): e106960, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25310187

RESUMEN

Chronic stress or long-term administration of glucocorticoids disrupts the hypothalamus-pituitary-adrenal system leading to continuous high levels of glucocorticoids and insulin resistance (IR). This pre-diabetic state can eventually develop into type 2 diabetes mellitus and has been associated with a higher risk to develop depressive disorders. The mechanisms underlying the link between chronic stress, IR and depression remains unclear. The present study aimed to establish a stress-depression model in mice to further study the effects of stress-induced changes upon insulin sensitivity and behavioural consequences. A pilot study was conducted to establish the optimal administration route and a pragmatic measurement of IR. Subsequently, 6-month-old C57BL/6NCrl mice were exposed to long-term oral corticosterone treatment via the drinking water. To evaluate insulin sensitivity changes, blood glucose and plasma insulin levels were measured at different time-points throughout treatment and mice were behaviourally assessed in the elevated zero maze (EZM), forced swimming test (FST) and open field test to reveal behavioural changes. Long-term corticosterone treatment increased body weight and decreased insulin sensitivity. The latter was revealed by a higher IR index and increased insulin in the plasma, whereas blood glucose levels remained unchanged. Corticosterone treatment induced longer immobility times in the FST, reflecting depressive-like behaviour. No effects were observed upon anxiety as measured in the EZM. The effect of the higher body weight of the CORT treated animals at time of testing did not influence behaviour in the EZM or FST, as no differences were found in general locomotor activity. Long-term corticosterone treatment via the drinking water reduces insulin sensitivity and induces depressive-like behaviour in the C57BL/6 mouse. This mouse model could thus be used to further explore the underlying mechanisms of chronic stress-induced T2DM and its association with increased prevalence of major depressive disorder on the short-term and other behavioural adaptations on the longer term.


Asunto(s)
Conducta Animal/efectos de los fármacos , Corticosterona/farmacología , Depresión/inducido químicamente , Resistencia a la Insulina/fisiología , Insulina/sangre , Animales , Conducta Animal/fisiología , Glucemia , Depresión/sangre , Depresión/fisiopatología , Modelos Animales de Enfermedad , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proyectos Piloto , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/fisiopatología , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología , Natación
15.
Neuropharmacology ; 64: 124-36, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22771768

RESUMEN

Alzheimer's disease (AD) is characterized by progressive cognitive deficits and synaptic dysfunction. Over the last decade phosphodiesterase inhibitors (PDEIs) have received increasing attention as putative cognition enhancers and have been suggested as a novel treatment strategy for AD. Given their ability to prevent hydrolysis of cAMP and/or cGMP, they can stimulate the cAMP/protein kinase A (PKA)/cAMP element-binding protein (CREB) and cGMP/PKG/CREB pathway to enhance synaptic transmission by increasing CREB phosphorylation (pCREB) and brain-derived neurotrophic factor (BDNF) transcription. Based on previous research, we hypothesized that chronic PDE2I treatment would improve AD-related cognitive deficits, by decreasing amyloid-ß (Aß) plaque load, enhancing pCREB and BDNF levels and increasing synaptic density in the hippocampus of 8-month-old APPswe/PS1dE9 mice. Results indicated that chronic PDE2I treatment could indeed improve memory performance in APPswe/PS1dE9 mice, without affecting anxiety, depressive-like behavior or hypothalamus-pituitary-adrenal axis regulation. However, no treatment effects were observed on Aß plaque load, pCREB or BDNF concentrations, or presynaptic density in the hippocampus, suggesting that other signaling pathways and/or effector molecules might be responsible for its cognition-enhancing effects. Presynaptic density in the stratum lucidum of the CA3 subregion was significantly higher in APPswe/PS1dE9 mice compared to WT controls, possibly reflecting a compensatory mechanism. In conclusion, PDEs in general, and PDE2 specifically, could be considered as promising therapeutic targets for cognition enhancement in AD, although the underlying mechanism of action remains to be elucidated. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Modelos Animales de Enfermedad , Trastornos de la Memoria/prevención & control , Memoria a Corto Plazo/efectos de los fármacos , Nootrópicos/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/prevención & control , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Imidazoles/uso terapéutico , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Distribución Aleatoria , Reconocimiento en Psicología/efectos de los fármacos , Conducta Espacial/efectos de los fármacos , Triazinas/uso terapéutico
16.
Neurobiol Aging ; 34(1): 319-37, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22738723

RESUMEN

Epidemiological evidence implies a role for chronic stress and stress-related disorders in the etiopathogenesis of sporadic Alzheimer's disease (AD). Although chronic stress exposure during various stages of life has been shown to exacerbate AD-related cognitive deficits and neuropathology in AD mouse models, the role of stress exposure during the prenatal period on AD development and progression remained to be investigated. The present study therefore explored the effects of prenatal maternal stress (PMS) in both male and female APPswe/PS1dE9 mouse offspring in terms of cognition, affect, and AD-related neuropathology. As prenatal perturbations are likely to mediate their effects via alterations in epigenetic regulation, changes in hippocampal DNA methyltransferase 3a, 5-methylcytosine and 5-hydroxymethylcytosine levels were assessed as underlying mechanisms. Repetitive restraint stress during the first week of gestation exerted a sex-dependent effect, with male PMS mice showing spatial memory deficits and a blunted hypothalamus-pituitary-adrenal axis response, while female PMS mice showed improved spatial memory performance, increased depressive-like behavior, as well as a decrease in hippocampal plaque load. In addition, sex differences were observed among APPswe/PS1dE9 mice, independent of PMS (i.e., female mice showed impaired spatial memory performance, higher hippocampal plaque load, altered amyloid precursor protein processing in the CA3 and lower DNA methyltransferase 3a immunoreactivity in the dentate gyrus when compared with male mice of the same age). In conclusion, PMS exposure impacts on the behavioral phenotype and neuropathology of APPswe/PS1dE9 mice. Moreover, given the remarkable sex differences observed, one should not overlook the impact of sex-specific responses to environmental exposures when investigating gene-environment interactions in AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Síntomas Conductuales/etiología , Trastornos del Conocimiento/etiología , Trastornos de la Memoria/etiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Estrés Psicológico/complicaciones , 5-Metilcitosina/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Benzofuranos , Citosina/análogos & derivados , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Femenino , Hipocampo/enzimología , Humanos , Masculino , Ratones , Ratones Transgénicos , Embarazo , Presenilina-1/genética , Quinolinas , Percepción Espacial/fisiología , Estrés Psicológico/patología
17.
EMBO Mol Med ; 5(10): 1613-34, 2013 10.
Artículo en Inglés | MEDLINE | ID: mdl-24014289

RESUMEN

An overview of miRNAs altered in Alzheimer's disease (AD) was established by profiling the hippocampus of a cohort of 41 late-onset AD (LOAD) patients and 23 controls, showing deregulation of 35 miRNAs. Profiling of miRNAs in the prefrontal cortex of a second independent cohort of 49 patients grouped by Braak stages revealed 41 deregulated miRNAs. We focused on miR-132-3p which is strongly altered in both brain areas. Downregulation of this miRNA occurs already at Braak stages III and IV, before loss of neuron-specific miRNAs. Next-generation sequencing confirmed a strong decrease of miR-132-3p and of three family-related miRNAs encoded by the same miRNA cluster on chromosome 17. Deregulation of miR-132-3p in AD brain appears to occur mainly in neurons displaying Tau hyper-phosphorylation. We provide evidence that miR-132-3p may contribute to disease progression through aberrant regulation of mRNA targets in the Tau network. The transcription factor (TF) FOXO1a appears to be a key target of miR-132-3p in this pathway.


Asunto(s)
Enfermedad de Alzheimer/genética , MicroARNs/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Área Bajo la Curva , Encéfalo/metabolismo , Cromosomas Humanos Par 17 , Análisis por Conglomerados , Estudios de Cohortes , Progresión de la Enfermedad , Regulación hacia Abajo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Hipocampo/metabolismo , Humanos , Neuronas/metabolismo , Fosforilación , Curva ROC , Índice de Severidad de la Enfermedad , Proteínas tau/metabolismo
18.
Neurochem Int ; 61(5): 697-701, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22750275

RESUMEN

Chronic stress and stress-related disorders, such as major depression (MD), have been shown to increase the risk for developing Alzheimer's disease (AD). Brain-derived neurotrophic factor (BDNF) has been postulated as a neurophysiological link between these illnesses. Our previous research has indicated that exposing the APPswe/PS1dE9 mouse model of AD to prenatal maternal stress (PS) induced a depressive-like phenotype, specifically in female mice. Considering the role of BDNF in depressive-like behavior and its interactions with amyloid-ß (Aß), our aim was to explore whether these mice would also exhibit alterations in soluble Aß, mature BDNF (mBDNF), proBDNF, and the receptors TrkB and p75(NTR) in comparison to non-stressed animals. Our results demonstrate that female APPswe/PS1dE9 mice have higher levels of hippocampal proBDNF and soluble Aß as compared to their male littermates. Additionally, a tendency was observed for PS to lower mBDNF protein levels in the hippocampus, but only in female mice, while receptor levels remained unaltered by sex or PS exposure. Given that female mice both have higher proBDNF and Aß levels, these findings suggest an underlying role for BDNF signaling and Aß production in the selective vulnerability of women for MD and AD development.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fragmentos de Péptidos/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Caracteres Sexuales , Estrés Psicológico/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Presenilina-1/genética , Solubilidad , Estrés Psicológico/genética
19.
Brain Struct Funct ; 216(3): 227-37, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21409417

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by amyloid plaque accumulation, intracellular tangles and neuronal loss in selective brain regions. The frontal cortex, important for executive functioning, is one of the regions that are affected. Here, we investigated the neurodegenerative effects of mutant human amyloid precursor protein (APP) and presenilin 1 (PS1) on frontal cortex neurons in APP/PS1KI mice, a transgenic mouse model of AD, expressing two mutations in the human APP, as well as two human PS1 mutations knocked-in into the mouse PS1 gene in a homozygous (ho) manner. Although the hippocampus is significantly affected in these mice, very little is known about the effects of these mutations on selective neuronal populations and plaque load in the frontal cortex. In this study, cytoarchitectural changes were characterized using high precision design-based stereology to evaluate plaque load, total neuron numbers, as well as total numbers of parvalbumin- (PV) and calretinin- (CR) immunoreactive (ir) neurons in the frontal cortex of 2- and 10-month-old APP/PS1KI mice. The frontal cortex was divided into two subfields: layers II-IV and layers V-VI, the latter of which showed substantially more extracellular amyloid-beta aggregates. We found a 34% neuron loss in layers V-VI in the frontal cortex of 10-month-old APP/PS1KI mice compared to 2-month-old, while there was no change in PV- and CR-ir neurons in these mice. In addition, the plaque load in layers V-VI of 10-month-old APP/PS1KI mice was only 11% and did not fully account for the extent of neuronal loss. Interestingly, an increase was found in the total number of PV-ir neurons in all frontal cortical layers of single transgenic APP mice and in layers II-IV of single transgenic PS1ho mice between 2 and 10 months of age. In conclusion, the APP/PS1KI mice provide novel insights into the regional selective vulnerability in the frontal cortex during AD that, together with previous findings in the hippocampus, are remarkably similar to the human situation.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Lóbulo Frontal/patología , Degeneración Nerviosa/patología , Neuronas/patología , Presenilina-1/metabolismo , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Técnicas de Sustitución del Gen , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mutación/genética , Degeneración Nerviosa/etiología , Presenilina-1/genética
20.
Eur J Pharmacol ; 626(1): 72-82, 2010 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-19837053

RESUMEN

Major depression (MD) is a severe mental disorder characterized by alterations in mood and cognition, with disease severity correlating inversely with cognition scores. Neuropathology can be found abundantly in the limbic system, which is thought to regulate affect, attention and memory. Hypothalamic-pituitary-adrenal (HPA) axis overdrive, as well as decreased serotonin levels, have often been implicated in the pathogenesis of this illness. Interestingly, there is substantial interaction between these two systems, with receptors of one system influencing the function of the other. This results in impaired neural networks, which give rise to the wide range of depressive symptoms. Recently, it has been implied that MD could serve as a risk factor for developing Alzheimer's disease (AD), with patients suffering from lifetime depression having a twofold higher chance of developing AD and exhibiting more AD-related neuropathology. Modifications in the HPA-axis and the serotonergic system may contribute to the development of cognitive decline and eventually AD. These two systems may therefore be involved in the pathogenesis of both illnesses and could provide a link between MD and AD. Obtaining more knowledge on their interactive role in the relation between MD and AD may eventually aid in the development of more effective treatment strategies.


Asunto(s)
Enfermedad de Alzheimer , Trastornos del Conocimiento , Trastorno Depresivo Mayor , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Trastorno Depresivo Mayor/complicaciones , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Trastorno Depresivo Mayor/fisiopatología , Glándulas Endocrinas/metabolismo , Glándulas Endocrinas/fisiopatología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Neurotransmisores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA