Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain ; 147(5): 1899-1913, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38242545

RESUMEN

Aberrant cholesterol metabolism causes neurological disease and neurodegeneration, and mitochondria have been linked to perturbed cholesterol homeostasis via the study of pathological mutations in the ATAD3 gene cluster. However, whether the cholesterol changes were compensatory or contributory to the disorder was unclear, and the effects on cell membranes and the wider cell were also unknown. Using patient-derived cells, we show that cholesterol perturbation is a conserved feature of pathological ATAD3 variants that is accompanied by an expanded lysosome population containing membrane whorls characteristic of lysosomal storage diseases. Lysosomes are also more numerous in Drosophila neural progenitor cells expressing mutant Atad3, which exhibit abundant membrane-bound cholesterol aggregates, many of which co-localize with lysosomes. By subjecting the Drosophila Atad3 mutant to nutrient restriction and cholesterol supplementation, we show that the mutant displays heightened cholesterol dependence. Collectively, these findings suggest that elevated cholesterol enhances tolerance to pathological ATAD3 variants; however, this comes at the cost of inducing cholesterol aggregation in membranes, which lysosomal clearance only partly mitigates.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas , Colesterol , Lisosomas , Proteínas de la Membrana , Mutación , Animales , Colesterol/metabolismo , Humanos , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Drosophila , Membrana Celular/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(4): 991-1001, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28132883

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays pivotal roles in neuronal function. The cleaved - mature - form of BDNF (mBDNF), predominantly expressed in adult brains, critically determines its effects. However, insufficient proteolytic processing under pathology may lead to the precursor form of BDNF (proBDNF) and thereby increased neuronal apoptosis and synaptic weakening. Previous findings in our lab showed that cognitive stimulation (CS) delayed memory decline in Tg2576 mouse model of Alzheimer's disease (AD), an effect that was tightly associated with augmented levels of mBDNF. In view of this association, the present study explored whether altered cleavage of BDNF could be involved in AD-related traits triggered by excessive amyloid-ß (Aß) pathology and whether this process could be therapeutically targeted. Aß pathology, both in AD patient samples and experimental models, triggered the upregulation of plasminogen-activator inhibitor-1 (PAI-1) via JNK/c-Jun. This led to inhibition of plasmin-regulated conversion of mBDNF. Pharmacological inhibition of PAI-1 with PAI-039 sufficiently reverted Aß-induced tau hyperphosphorylation and neurotoxicity. Chronic treatment of 15 old-month Tg2576 mice with oral administration of PAI-039 resulted in improved BDNF maturation and cognitive function without inducing significant changes in amyloid burden. In conclusion, upregulation of PAI-1 may be a critical mechanism underlying insufficient neurotrophic support and increased neurodegeneration associated with AD. Thus, targeting BDNF maturation through pharmacological inhibition of PAI-1 might become a potential treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Serpina E2/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Humanos , Ácidos Indolacéticos/farmacología , Ratones , Ratones Transgénicos , Inhibidor 1 de Activador Plasminogénico/genética , Serpina E2/genética
3.
Hippocampus ; 26(10): 1303-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27258819

RESUMEN

Alzheimer's disease (AD) is characterized phenotypically by memory impairment, histologically by accumulation of pTau and ß-amyloid peptide and morphologically by a loss of nerve terminals in cortical and hippocampal regions. As glutamate is the principle excitatory neurotransmitter of the central nervous system (CNS), the glutamatergic system may play an important role in AD. To date, not many studies have addressed the deleterious effects of Aß on glutamatergic terminals; therefore the aim of this study was to investigate how Aß affects glutamatergic terminals and to assess the extent to which alterations in the glutamatergic neurotransmission could impact susceptibility to the illness. The present study shows that Aß caused a loss of glutamatergic terminals, measured by VGLUT1 protein levels, in Tg2576 primary cell cultures, Tg2576 mice and AD patient brains, and also when Aß was added exogenously to hippocampal cell cultures. Interestingly, no correlation was found between cognition and decreased VGLUT1 levels. Moreover, when Aß1-42 was intracerebroventricularlly administered into VGLUT1+/- mice, altered synaptic plasticity and increased neuroinflammation was observed in the hippocampus of those animals. In conclusion, the present study not only revealed susceptibility of glutamatergic nerve terminals to Aß induced toxicity but also underlined the importance of VGLUT1 in the progression of AD, as the decrease of this protein levels could increase the susceptibility to subsequent deleterious inputs by exacerbating Aß induced neuroinflammation and synaptic plasticity disruption. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Neuronas/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/genética
4.
J Biol Chem ; 286(29): 25574-85, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21628469

RESUMEN

Disturbances in cholesterol metabolism have been associated with hypertension and neurodegenerative disorders. Because cholesterol metabolism in the brain is efficiently separated from plasma cholesterol by the blood-brain barrier (BBB), it is an unsolved paradox how high blood cholesterol can cause an effect in the brain. Here, we discuss the possibility that cholesterol metabolites permeable to the BBB might account for these effects. We show that 27-hydroxycholesterol (27-OH) and 24S-hydroxycholesterol (24S-OH) up-regulate the renin-angiotensin system (RAS) in the brain. Brains of mice on a cholesterol-enriched diet showed up-regulated angiotensin converting enzyme (ACE), angiotensinogen (AGT), and increased JAK/STAT activity. These effects were confirmed in in vitro studies with primary neurons and astrocytes exposed to 27-OH or 24S-OH, and were partially mediated by liver X receptors. In contrast, brain RAS activity was decreased in Cyp27a1-deficient mice, a model exhibiting reduced 27-OH production from cholesterol. Moreover, in humans, normocholesterolemic patients with elevated 27-OH levels, due to a CYP7B1 mutation, had markers of activated RAS in their cerebrospinal fluid. Our results demonstrate that side chain-oxidized oxysterols are modulators of brain RAS. Considering that levels of cholesterol and 27-OH correlate in the circulation and 27-OH can pass the BBB into the brain, we suggest that this cholesterol metabolite could be a link between high plasma cholesterol levels, hypertension, and neurodegeneration.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hidroxicolesteroles/farmacología , Receptores Nucleares Huérfanos/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Angiotensinógeno/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Colesterol en la Dieta/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Hidroxicolesteroles/química , Hidroxicolesteroles/metabolismo , Receptores X del Hígado , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Ratas , Paraplejía Espástica Hereditaria/líquido cefalorraquídeo , Paraplejía Espástica Hereditaria/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Front Cell Dev Biol ; 10: 836196, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35419363

RESUMEN

Neurons are post-mitotic cells that allocate huge amounts of energy to the synthesis of new organelles and molecules, neurotransmission and to the maintenance of redox homeostasis. In neurons, autophagy is not only crucial to ensure organelle renewal but it is also essential to balance nutritional needs through the mobilization of internal energy stores. A delicate crosstalk between the pathways that sense nutritional status of the cell and the autophagic processes to recycle organelles and macronutrients is fundamental to guarantee the proper functioning of the neuron in times of energy scarcity. This review provides a detailed overview of the pathways and processes involved in the balance of cellular energy mediated by autophagy, which when defective, precipitate the neurodegenerative cascade of Parkinson's disease, frontotemporal dementia, amyotrophic lateral sclerosis or Alzheimer's disease.

6.
Br J Pharmacol ; 178(6): 1269-1278, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32497246

RESUMEN

Amyotrophic lateral sclerosis (ALS) commonly referred to as motor neurone disease, is a neurodegenerative disease of unknown pathogenesis that progresses rapidly and has attracted an increased amount of scholarly interest in recent years. The current conception of amyotrophic lateral sclerosis has transitioned into a more complex theory in which individual genetic risk, ageing and environmental factors interact, leading to disease onset in subjects in whom the sum of these factors reach a determined threshold. Based on this conceptualization, the environmental conditions, particularly those that are potentially modifiable, are becoming increasingly relevant. In this review, the current integrative model of the disease is discussed. In addition, we explore the role of cancer, autoimmunity and metabolic diseases as examples of novel, non-genetic and environmental factors. Together with the potential triggers or perpetuating pathogenic mechanisms along with new insights into potential lines of future research are provided. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.


Asunto(s)
Esclerosis Amiotrófica Lateral , Neoplasias , Enfermedades Neurodegenerativas , Autoinmunidad , Humanos , Factores de Riesgo
7.
J Pers Med ; 11(7)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34357138

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.

8.
Artículo en Inglés | MEDLINE | ID: mdl-32106710

RESUMEN

Objective: To perform a comprehensive lipid profiling to evaluate potential lipid metabolic differences between patients with amyotrophic lateral sclerosis (ALS) and controls, and to provide a more profound understanding of the metabolic abnormalities in ALS. Methods: Twenty patients with ALS and 20 healthy controls were enrolled in a cross-sectional study. Untargeted lipidomics profiling in fasting serum samples were performed by optimized UPLC-MS platforms for broad lipidome coverage. Datasets were analyzed by univariate and a variety of multivariate procedures. Results: We provide the most comprehensive blood lipid profiling of ALS to date, with a total of 416 lipids measured. Univariate analysis showed that 28 individual lipid features and two lipid classes, triacylglycerides and oxidized fatty acids (FAs), were altered in patients with ALS, although none of these changes remained significant after multiple comparison adjustment. Most of these changes remained constant after removing from the analysis individuals treated with lipid-lowering drugs. The non-supervised principal component analysis did not identify any lipid clustering of patients with ALS and controls. Despite this, we performed a variety of linear and non-linear supervised multivariate models to select the most reliable features that discriminate the lipid profile of patients with ALS from controls. These were the monounsaturated FAs C24:1n-9 and C14:1, the triglyceride TG(51:4) and the sphingomyelin SM(36:2). Conclusions: Peripheral alterations of lipid metabolism are poorly defined in ALS, triacylglycerides and certain types of FAs could contribute to the different lipid profile of patients with ALS. These findings should be validated in an independent cohort.


Asunto(s)
Esclerosis Amiotrófica Lateral/sangre , Esclerosis Amiotrófica Lateral/diagnóstico , Lipidómica/métodos , Espectrometría de Masas en Tándem/métodos , Anciano , Cromatografía Líquida de Alta Presión/métodos , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad
9.
Prog Neurobiol ; 142: 104-129, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27236050

RESUMEN

The scientific scenario of amyotrophic lateral sclerosis (ALS) has dramatically changed since TDP-43 aggregates were discovered in 2006 as the main component of the neuronal inclusions seen in the disease, and more recently, when the implication of C9ORF72 expansion in familial and sporadic cases of ALS and frontotemporal dementia was confirmed. These discoveries have enlarged an extense list of genes implicated in different cellular processes such as RNA processing or autophagia among others and have broaden the putative molecular targets of the disease. Some of ALS-related genes such as TARDBP or SOD1 among others have important roles in the regulation of glucose and fatty acids metabolism, so that an impairment of fatty acids (FA) consumption and ketogenic deficits during exercise in ALS patients would connect the physiopathology with some of the more intriguing epidemiological traits of the disease. The current understanding of ALS as part of a continuum with other neurodegenerative diseases and a crossroads between genetic, neurometabolic and environmental factors represent a fascinating model of interaction that could be translated to other neurodegenerative diseases. In this review we summarize the most relevant data obtained in the ten last years and the key lines for future research in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Interacción Gen-Ambiente , Esclerosis Amiotrófica Lateral/etiología , Animales , Humanos
10.
Neurosci Lett ; 375(1): 37-41, 2005 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-15664119

RESUMEN

Cognitive deficits in neuropsychiatric disorders, such as Alzheimer's disease (AD), have been closely related to cholinergic deficits. We have compared different markers of cholinergic function to assess the best biomarker of cognitive deficits associated to cholinergic hypoactivity. In post-mortem frontal cortex from AD patients, acetylcholine (ACh) levels, cholinacetyltransferase (ChAT) and acetylcholinesterase (AChE) activity were all reduced compared to controls. Both ChAT and AChE activity showed a significant correlation with cognitive deficits. In the frontal cortex of rats with a selective cholinergic lesion, all cholinergic parameters measured (ACh levels, ChAT and AChE activities, "in vitro" and "in vivo" basal ACh release) were significantly reduced. AChE activity was associated to ChAT activity, and even more, to "in vivo" and "in vitro" basal ACh release. Quantification of AChE activity is performed by an easy and cheap method and therefore, these results suggest that determination of AChE activity may be used as an effective first step method to evaluate cholinergic deficits.


Asunto(s)
Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Sulfatasas/metabolismo , Anciano , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/fisiopatología , Análisis de Varianza , Animales , Anticuerpos Monoclonales , Biomarcadores/metabolismo , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunotoxinas , Masculino , Escala del Estado Mental , N-Glicosil Hidrolasas , Cambios Post Mortem , Ratas , Proteínas Inactivadoras de Ribosomas Tipo 1 , Saporinas , Estadística como Asunto , Factores de Tiempo
11.
Neurosci Lett ; 506(2): 245-50, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22119000

RESUMEN

Estrogen was shown to promote neuronal survival against several neurotoxic insults including ß-amyloid (Aß). The proposed mechanism includes the activation of the mitogen activated protein kinase/extracellular signal-regulated kinase (Mapk/Erk), phosphatidylinositol 3-kinase/Akt pathways and the upregulation of antiapoptotic proteins. On the other hand, Aß neurotoxicity depends on the activation of apoptosis signal-regulating kinase 1 (Ask1), and both Ask1 activity and Aß toxicity are inhibited by thioredoxin-1 (Trx1). Here, we explored the possibility that estrogen could protect cells against Aß(1-42) toxicity by inhibiting the Ask1 cascade or by modulating Trx1. Cytosolic translocation of death-associated protein Daxx was used as indicator of Ask1 activity. Using human SH-SY5Y neuroblastoma cells, 17ß-estradiol (E2) and specific agonists for estrogen receptor (ER) α or ß we demonstrated that nM concentrations of E2 protected against Aß(1-42) by a mechanism depending upon ERα stimulation, Akt activation and Ask1 inhibition. Moreover, this protection would occur independently of ERß and the induction of Trx1 expression. Our results emphasize the importance of Ask1 cascade in Aß toxicity, and of ERα and Ask1 as targets for developing new neuroprotective drugs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Péptidos beta-Amiloides/toxicidad , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteínas Nucleares/genética , Línea Celular Tumoral , Proteínas Co-Represoras , Humanos , Inmunohistoquímica , Chaperonas Moleculares , Fármacos Neuroprotectores/farmacología , Translocación Genética
12.
J Alzheimers Dis ; 24(4): 669-79, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21297254

RESUMEN

In spite of the fact that cholesterol does not pass the blood-brain barrier, hypercholesterolemia has been linked to increase Alzheimer's disease (AD) risk. Hypertension is another risk factor and angiotensin converting enzyme (ACE) activity is known to be increased in AD. Furthermore, a lower incidence of AD has been reported in patients taking anti-hypertensive drugs. Here we show that the levels of angiotensinogen (AGT) and ACE are increased in the cerebrospinal fluid (CSF) of patients with mild cognitive impairment and AD. Moreover, we show ACE activity in the CSF to be positively correlated with both plasma and CSF levels of 27-hydroxycholesterol (27-OH), an oxysterol known to pass through the BBB and taken up from the circulation by the brain. In addition, treatment of rat primary neurons, astrocytes, and human neuroblastoma cells with 27-OH resulted in increased production of AGT. Our results demonstrate that upregulation of renin-angiotensin system (RAS) in AD brains occurs not only at the enzymatic level (ACE) but also at the substrate level (AGT). The possibility that 27-OH is part of a mechanism linking hypercholesterolemia with increased brain RAS activity and increased AD risk is discussed.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Química Encefálica/fisiología , Disfunción Cognitiva/metabolismo , Hidroxicolesteroles/química , Hidroxicolesteroles/metabolismo , Sistema Renina-Angiotensina/fisiología , Regulación hacia Arriba/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Angiotensinógeno/biosíntesis , Angiotensinógeno/fisiología , Animales , Biomarcadores/metabolismo , Química Encefálica/efectos de los fármacos , Células Cultivadas , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/fisiopatología , Estudios Transversales , Femenino , Humanos , Hidroxicolesteroles/farmacología , Masculino , Persona de Mediana Edad , Peptidil-Dipeptidasa A/biosíntesis , Peptidil-Dipeptidasa A/fisiología , Ratas , Ratas Sprague-Dawley , Sistema Renina-Angiotensina/efectos de los fármacos
13.
Cell Metab ; 14(2): 242-53, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21803294

RESUMEN

Cardiotrophin-1 (CT-1) is a member of the gp130 family of cytokines. We observed that ct-1(-/-) mice develop mature-onset obesity, insulin resistance, and hypercholesterolemia despite reduced calorie intake. Decreased energy expenditure preceded and accompanied the development of obesity. Acute treatment with rCT-1 decreased blood glucose in an insulin-independent manner and increased insulin-stimulated AKT phosphorylation in muscle. These changes were associated with stimulation of fatty acid oxidation, an effect that was absent in AMPKα2(-/-) mice. Chronic rCT-1 treatment reduced food intake, enhanced energy expenditure, and induced white adipose tissue remodeling characterized by upregulation of genes implicated in the control of lipolysis, fatty acid oxidation, and mitochondrial biogenesis and genes typifying brown fat phenotype. Moreover, rCT-1 reduced body weight and corrected insulin resistance in ob/ob and in high-fat-fed obese mice. We conclude that CT-1 is a master regulator of fat and glucose metabolism with potential applications for treatment of obesity and insulin resistance.


Asunto(s)
Citocinas/metabolismo , Glucosa/metabolismo , Metabolismo de los Lípidos , Tejido Adiposo Blanco/metabolismo , Animales , Peso Corporal , Citocinas/deficiencia , Citocinas/farmacología , Ingestión de Alimentos , Metabolismo Energético , Ácidos Grasos/metabolismo , Hipercolesterolemia/genética , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Oxidación-Reducción , Fosforilación
14.
Psychoneuroendocrinology ; 34(10): 1495-505, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19505767

RESUMEN

Adverse experiences early in life may sensitize specific neurocircuits to subsequent stressors. We have evaluated in maternal separation (MS) rats, an animal paradigm of early-life stress, the effects of a selective cholinergic lesion on cognitive function as well as susceptibility of cholinergic neurons to the lesion. MS rats subjected to a cholinergic lesion by administration of the immunotoxin 192 IgG-saporin, showed significant decreases in both choline acetyltransferase (ChAT) and acetylcholinesterase (AChE) activity compared to control lesioned rats. Morris water maze results revealed a significant impairment in learning and memory function in MS adult rats and further cognitive deficits after the lesion. The lesion of cholinergic neurons induced a significant decrease in glucocorticoid receptor density in MS rats, accompanied by increases in CRF mRNA expression. Decreases in NGF and increases in NGF-p75NTR expression have also been found in MS rats. Our results suggest that vulnerability of basal forebrain cholinergic nerve cells might be affected by the HPA axis. The present data are discussed not only in terms of conditions that occur during ageing or Alzheimer disease, but also regarding a purported involvement of the cholinergic system in the regulation of HPA axis activity.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Fibras Colinérgicas/metabolismo , Cognición/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Inmunotoxinas/inmunología , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Animales , Animales Recién Nacidos , Corteza Cerebral/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Privación Materna , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores de Glucocorticoides/metabolismo , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/metabolismo , Saporinas
15.
Brain Pathol ; 19(1): 69-80, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18503570

RESUMEN

Growing evidence strongly suggests that high fat diet (HFD) has an important role in some neurodegenerative disorders, including Alzheimer's disease (AD). To identify new cellular pathways linking hypercholesterolemia and neurodegeneration, we analyzed the effects of HFD on gene expression in mouse brain. Using cDNA microarrays and real time RT-PCR, we found that HFD has a mild, but significant effect on the expression of several genes. The altered genes include molecules linked to AD pathology and others of potential interest for neurodegeneration. We further investigated the effect of HFD on the activity-regulated cytoskeleton-associated protein (Arc). Expression of Arc was decreased in cerebral cortex and hippocampus of HFD-fed animals. From the known regulatory mechanisms of Arc expression, HFD reduced N-methyl-D-aspartate receptor (NMDAR) activity, as seen by decreases in tyrosine phosphorylation of NMDAR2A and levels of NMDAR1. Additionally, we demonstrated that 27-hydroxycholesterol, a cholesterol metabolite that enters the brain from the blood, decreases Arc levels as well as NMDAR and Src kinase activities in rat primary hippocampal neurons. Finally, we showed that Arc levels are decreased in the cortex of AD brains. We propose that one of the mechanisms, by which hypercholesterolemia contributes to neurodegenerative diseases, could be through Arc down-regulation caused by 27-hydroxycholesterol.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Grasas de la Dieta/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Arginina Vasopresina/genética , Encéfalo/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Grasas de la Dieta/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hidroxicolesteroles/farmacología , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/genética , ARN/metabolismo , Receptores de Glutamato/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA