Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(10)2021 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065232

RESUMEN

Tau protein is largely responsible for tauopathies, including Alzheimer's disease (AD), where it accumulates in the brain as insoluble aggregates. Tau mRNA is regulated by alternative splicing, and inclusion or exclusion of exon 10 gives rise to the 3R and 4R isoforms respectively, whose balance is physiologically regulated. In this sense, one of the several factors that regulate alternative splicing of tau is GSK3ß, whose activity is inhibited by the cellular prion protein (PrPC), which has different physiological functions in neuroprotection and neuronal differentiation. Moreover, a relationship between PrPC and tau expression levels has been reported during AD evolution. For this reason, in this study we aimed to analyze the role of PrPC and the implication of GSK3ß in the regulation of tau exon 10 alternative splicing. We used AD human samples and mouse models of PrPC ablation and tau overexpression. In addition, we used primary neuronal cultures to develop functional studies. Our results revealed a paralleled association between PrPC expression and tau 4R isoforms in all models analyzed. In this sense, reduction or ablation of PrPC levels induces an increase in tau 3R/4R balance. More relevantly, our data points to GSK3ß activity downstream from PrPC in this phenomenon. Our results indicate that PrPC plays a role in tau exon 10 inclusion through the inhibitory capacity of GSK3ß.


Asunto(s)
Regulación hacia Abajo/genética , Exones/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Priones/genética , Proteínas tau/genética , Adulto , Anciano , Anciano de 80 o más Años , Empalme Alternativo/genética , Enfermedad de Alzheimer/genética , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Cuerpos de Inclusión/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neuronas/patología , Isoformas de Proteínas/genética , ARN Mensajero/genética , Tauopatías/genética
2.
Cell Mol Life Sci ; 72(14): 2719-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25708702

RESUMEN

Olfactory ensheathing cell (OEC) transplantation emerged some years ago as a promising therapeutic strategy to repair injured spinal cord. However, inhibitory molecules are present for long periods of time in lesioned spinal cord, inhibiting both OEC migration and axonal regrowth. Two families of these molecules, chondroitin sulphate proteoglycans (CSPG) and myelin-derived inhibitors (MAIs), are able to trigger inhibitory responses in lesioned axons. Mounting evidence suggests that OEC migration is inhibited by myelin. Here we demonstrate that OEC migration is largely inhibited by CSPGs and that inhibition can be overcome by the bacterial enzyme Chondroitinase ABC. In parallel, we have generated a stable OEC cell line overexpressing the Nogo receptor (NgR) ectodomain to reduce MAI-associated inhibition in vitro and in vivo. Results indicate that engineered cells migrate longer distances than unmodified OECs over myelin or oligodendrocyte-myelin glycoprotein (OMgp)-coated substrates. In addition, they also show improved migration in lesioned spinal cord. Our results provide new insights toward the improvement of the mechanisms of action and optimization of OEC-based cell therapy for spinal cord lesion.


Asunto(s)
Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Neuroglía/fisiología , Animales , Axones/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Proteoglicanos Tipo Condroitín Sulfato/farmacología , Clonación Molecular , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Técnicas Analíticas Microfluídicas , Proteínas de la Mielina/genética , Neuroglía/metabolismo , Receptor Nogo 1 , Bulbo Olfatorio/citología , Glicoproteína Oligodendrócito-Mielina/farmacología , Estructura Terciaria de Proteína , Ratas , Receptores de Superficie Celular/genética , Traumatismos de la Médula Espinal/terapia , Imagen de Lapso de Tiempo
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167187, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38653354

RESUMEN

Clinical relevance of miRNAs as biomarkers is growing due to their stability and detection in biofluids. In this, diagnosis at asymptomatic stages of Alzheimer's disease (AD) remains a challenge since it can only be made at autopsy according to Braak NFT staging. Achieving the objective of detecting AD at early stages would allow possible therapies to be addressed before the onset of cognitive impairment. Many studies have determined that the expression pattern of some miRNAs is dysregulated in AD patients, but to date, none has been correlated with downregulated expression of cellular prion protein (PrPC) during disease progression. That is why, by means of cross studies of miRNAs up-regulated in AD with in silico identification of potential miRNAs-binding to 3'UTR of human PRNP gene, we selected miR-519a-3p for our study. Then, in vitro experiments were carried out in two ways. First, we validated miR-519a-3p target on 3'UTR-PRNP, and second, we analyzed the levels of PrPC expression after using of mimic technology on cell culture. In addition, RT-qPCR was performed to analyzed miR-519a-3p expression in human cerebral samples of AD at different stages of disease evolution. Additionally, samples of other neurodegenerative diseases such as other non-AD tauopathies and several synucleinopathies were included in the study. Our results showed that miR-519a-3p overlaps with PRNP 3'UTR in vitro and promotes downregulation of PrPC. Moreover, miR-519a-3p was found to be up-regulated exclusively in AD samples from stage I to VI, suggesting its potential use as a novel label of preclinical stages of the disease.


Asunto(s)
Regiones no Traducidas 3' , Enfermedad de Alzheimer , Biomarcadores , MicroARNs , Proteínas Priónicas , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/diagnóstico , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Biomarcadores/metabolismo , Regiones no Traducidas 3'/genética , Femenino , Anciano , Masculino , Anciano de 80 o más Años , Proteínas PrPC/metabolismo , Proteínas PrPC/genética
4.
Eur J Med Chem ; 250: 115169, 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36753881

RESUMEN

A set of twenty-five thioxanthene-9-one and xanthene-9-one derivatives, that were previously shown to inhibit cholinesterases (ChEs) and amyloid ß (Aß40) aggregation, were evaluated for the inhibition of tau protein aggregation. All compounds exhibited a good activity, and eight of them (5-8, 10, 14, 15 and 20) shared comparable low micromolar inhibitory potency versus Aß40 aggregation and human acetylcholinesterase (AChE), while inhibiting human butyrylcholinesterase (BChE) even at submicromolar concentration. Compound 20 showed outstanding biological data, inhibiting tau protein and Aß40 aggregation with IC50 = 1.8 and 1.3 µM, respectively. Moreover, at 0.1-10 µM it also exhibited neuroprotective activity against tau toxicity induced by okadoic acid in human neuroblastoma SH-SY5Y cells, that was comparable to that of estradiol and PD38. In preliminary toxicity studies, these interesting results for compound 20 are somewhat conflicting with a narrow safety window. However, compound 10, although endowed with a little lower potency for tau and Aß aggregation inhibition additionally demonstrated good inhibition of ChEs and rather low cytotoxicity. Compound 4 is also worth of note for its high potency as hBChE inhibitor (IC50 = 7 nM) and for the three order of magnitude selectivity versus hAChE. Molecular modelling studies were performed to explain the different behavior of compounds 4 and 20 towards hBChE. The observed balance of the inhibitory potencies versus the relevant targets indicates the thioxanthene-9-one derivatives as potential MTDLs for AD therapy, provided that the safety window will be improved by further structural variations, currently under investigation.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Butirilcolinesterasa/metabolismo , Péptidos beta-Amiloides/metabolismo , Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/química , Estructura Molecular , Relación Estructura-Actividad , Neuroblastoma/tratamiento farmacológico , Diseño de Fármacos , Simulación del Acoplamiento Molecular
5.
J Neurochem ; 113(6): 1644-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20374426

RESUMEN

Lesioned axons do not regenerate in the adult mammalian CNS, owing to the over-expression of inhibitory molecules such as myelin-derived proteins or chondroitin sulphate proteoglycans. In order to overcome axon inhibition, strategies based on extrinsic and intrinsic treatments have been developed. For myelin-associated inhibition, blockage with NEP1-40, receptor bodies or IN-1 antibodies has been used. In addition, endogenous blockage of cell signalling mechanisms induced by myelin-associated proteins is a potential tool for overcoming axon inhibitory signals. We examined the participation of glycogen synthase kinase 3beta (GSK3beta) and extracellular-related kinase (ERK) 1/2 in axon regeneration failure in lesioned cortical neurons. We also investigated whether pharmacological blockage of GSK3beta and ERK1/2 activities facilitates regeneration after myelin-directed inhibition in two models: (i) cerebellar granule cells and (ii) lesioned entorhino-hippocampal pathway in slice cultures, and whether the regenerative effects are mediated by Nogo Receptor 1 (NgR1). We demonstrate that, in contrast to ERK1/2 inhibition, the pharmacological treatment of GSK3beta inhibition strongly facilitated regrowth of cerebellar granule neurons over myelin independently of NgR1. Finally, these regenerative effects were corroborated in the lesioned entorhino-hippocampal pathway in NgR1-/- mutant mice. These results provide new findings for the development of new assays and strategies to enhance axon regeneration in injured cortical connections.


Asunto(s)
Corteza Cerebral/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Neuritas/fisiología , Neuronas/citología , Aminofenoles/farmacología , Animales , Animales Recién Nacidos , Axotomía/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteínas Ligadas a GPI , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/citología , Humanos , Indoles/farmacología , Lisina/análogos & derivados , Lisina/metabolismo , Maleimidas/farmacología , Ratones , Ratones Noqueados , Mutación/genética , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/farmacología , Neuritas/efectos de los fármacos , Proteínas Nogo , Receptor Nogo 1 , Embarazo , Ratas , Receptores de Superficie Celular , Receptores de Péptidos/deficiencia , Factores de Tiempo , Versicanos/metabolismo
6.
Cells ; 9(3)2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32131451

RESUMEN

Cellular (also termed 'natural') prion protein has been extensively studied for many years for its pathogenic role in prionopathies after misfolding. However, neuroprotective properties of the protein have been demonstrated under various scenarios. In this line, the involvement of the cellular prion protein in neurodegenerative diseases other than prionopathies continues to be widely debated by the scientific community. In fact, studies on knock-out mice show a vast range of physiological functions for the protein that can be supported by its ability as a cell surface scaffold protein. In this review, we first summarize the most commonly described roles of cellular prion protein in neuroprotection, including antioxidant and antiapoptotic activities and modulation of glutamate receptors. Second, in light of recently described interaction between cellular prion protein and some amyloid misfolded proteins, we will also discuss the molecular mechanisms potentially involved in protection against neurodegeneration in pathologies such as Alzheimer's, Parkinson's, and Huntington's diseases.


Asunto(s)
Encéfalo/patología , Enfermedades del Sistema Nervioso/fisiopatología , Proteínas Priónicas/metabolismo , Anciano , Animales , Humanos , Ratones , Ratones Noqueados
7.
Mol Neurobiol ; 57(10): 4170-4186, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32683652

RESUMEN

Cellular prion protein (PrPC) is largely responsible for transmissible spongiform encephalopathies (TSEs) when it becomes the abnormally processed and protease resistant form PrPSC. Physiological functions of PrPC include protective roles against oxidative stress and excitotoxicity. Relevantly, PrPC downregulates tau levels, whose accumulation and modification are a hallmark in the advance of Alzheimer's disease (AD). In addition to the accumulation of misfolded proteins, in the initial stages of AD-affected brains display both increased reactive oxygen species (ROS) markers and levels of PrPC. However, the factors responsible for the upregulation of PrPC are unknown. Thus, the aim of this study was to uncover the different molecular actors promoting PrPC overexpression. In order to mimic early stages of AD, we used ß-amyloid-derived diffusible ligands (ADDLs) and tau cellular treatments, as well as ROS generation, to elucidate their particular roles in human PRNP promoter activity. In addition, we used specific chemical inhibitors and site-specific mutations of the PRNP promoter sequence to analyze the contribution of the main transcription factors involved in PRNP transcription under the analyzed conditions. Our results revealed that tau is a new modulator of PrPC expression independently of ADDL treatment and ROS levels. Lastly, we discovered that the JNK/c-jun-AP-1 pathway is involved in increased PRNP transcription activity by tau but not in the promoter response to ROS.


Asunto(s)
Proteínas Priónicas/metabolismo , Transcripción Genética , Proteínas tau/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Sitios de Unión , Células HEK293 , Humanos , Ligandos , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Priónicas/genética , Proteínas Priónicas/ultraestructura , Regiones Promotoras Genéticas/genética , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Solubilidad , Proteínas tau/ultraestructura
8.
Brain Pathol ; 30(2): 298-318, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31397930

RESUMEN

Human tau seeding and spreading occur following intracerebral inoculation of brain homogenates obtained from tauopathies in transgenic mice expressing natural or mutant tau, and in wild-type (WT) mice. The present study was geared to learning about the patterns of tau seeding, the cells involved and the characteristics of tau following intracerebral inoculation of homogenates from primary age-related tauopathy (PART: neuronal 4Rtau and 3Rtau), aging-related tau astrogliopathy (ARTAG: astrocytic 4Rtau) and globular glial tauopathy (GGT: 4Rtau with neuronal deposits and specific tau inclusions in astrocytes and oligodendrocytes). For this purpose, young and adult WT mice were inoculated unilaterally in the hippocampus or in the lateral corpus callosum with sarkosyl-insoluble fractions from PART, ARTAG and GGT cases, and were killed at variable periods of three to seven months. Brains were processed for immunohistochemistry in paraffin sections. Tau seeding occurred in the ipsilateral hippocampus and corpus callosum and spread to the septal nuclei, periventricular hypothalamus and contralateral corpus callosum, respectively. Tau deposits were mainly found in neurons, oligodendrocytes and threads; the deposits were diffuse or granular, composed of phosphorylated tau, tau with abnormal conformation and 3Rtau and 4Rtau independently of the type of tauopathy. Truncated tau at the aspartic acid 421 and ubiquitination were absent. Tau deposits had the characteristics of pre-tangles. A percentage of intracellular tau deposits co-localized with active (phosphorylated) tau kinases p38 and ERK 1/2. Present study shows that seeding and spreading of human tau into the brain of WT mice involves neurons and glial cells, mainly oligodendrocytes, thereby supporting the idea of a primary role of oligodendrogliopathy, together with neuronopathy, in the progression of tauopathies. In addition, it suggests that human tau inoculation modifies murine tau metabolism with the production and deposition of 3Rtau and 4Rtau, and by activation of specific tau kinases in affected cells.


Asunto(s)
Encéfalo/patología , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Cells ; 9(5)2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32438605

RESUMEN

Reelin is an extracellular glycoprotein that modulates neuronal function and synaptic plasticity in the adult brain. Decreased levels of Reelin activity have been postulated as a key factor during neurodegeneration in Alzheimer´s disease (AD) and in aging. Thus, changes in levels of full-length Reelin and Reelin fragments have been revealed in cerebrospinal fluid (CSF) and in post-mortem brains samples of AD patients with respect to non-AD patients. However, conflicting studies have reported decreased or unchanged levels of full-length Reelin in AD patients compared to control (nND) cases in post-mortem brains and CSF samples. In addition, a compelling analysis of Reelin levels in neurodegenerative diseases other than AD is missing. In this study, we analyzed brain levels of RELN mRNA and Reelin protein in post-mortem frontal cortex samples from different sporadic AD stages, Parkinson's disease with dementia (PDD), and Creutzfeldt-Jakob disease (sCJD), obtained from five different Biobanks. In addition, we measured Reelin protein levels in CSF samples of patients with mild cognitive impairment (MCI), dementia, or sCJD diagnosis and a group of neurologically healthy cases. The results indicate an increase in RELN mRNA in the frontal cortex of advanced stages of AD and in sCJD(I) compared to controls. This was not observed in PDD and early AD stages. However, Reelin protein levels in frontal cortex samples were unchanged between nND and advanced AD stages and PDD. Nevertheless, they decreased in the CSF of patients with dementia in comparison to those not suffering with dementia and patients with MCI. With respect to sCJD, there was a tendency to increase in brain samples in comparison to nND and to decrease in the CSF with respect to nND. In conclusion, Reelin levels in CSF cannot be considered as a diagnostic biomarker for AD or PDD. However, we feel that the CSF Reelin changes observed between MCI, patients with dementia, and sCJD might be helpful in generating a biomarker signature in prodromal studies of unidentified dementia and sCJD.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Proteínas de la Matriz Extracelular/genética , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/genética , Serina Endopeptidasas/genética , Encéfalo/metabolismo , Encéfalo/patología , Moléculas de Adhesión Celular Neuronal/líquido cefalorraquídeo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/líquido cefalorraquídeo , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/líquido cefalorraquídeo , Enfermedades Neurodegenerativas/patología , Cambios Post Mortem , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Reelina , Serina Endopeptidasas/líquido cefalorraquídeo , Serina Endopeptidasas/metabolismo
10.
Lab Chip ; 8(11): 1896-905, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18941691

RESUMEN

The electrophysiological characterisation of cultured neurons is of paramount importance for drug discovery, safety pharmacology and basic research in the neurosciences. Technologies offering low cost, low technical complexity and potential for scalability towards high-throughput electrophysiology on in vitro neurons would be advantageous, in particular for screening purposes. Here we describe a plastic culture substrate supporting low-complexity multi-unit loose-patch recording and stimulation of developing networks while retaining manufacturability compatible with low-cost and large-scale production. Our hybrid polydimethylsilane (PDMS)-on-polystyrene structures include chambers (6 mm in diameter) and microchannels (25 microm x 3.7 microm x 1 mm) serving as substrate-embedded recording pipettes. Somas are plated and retained in the chambers due to geometrical constraints and their processes grow along the microchannels, effectively establishing a loose-patch configuration without human intervention. We demonstrate that off-the-shelf voltage-clamp, current-clamp and extracellular amplifiers can be used to record and stimulate multi-unit activity with the aid of our dishes. Spikes up to 50 pA in voltage-clamp and 300 microV in current-clamp modes are recorded in sparse and bursting activity patterns characteristic of 1 week-old hippocampal cultures. Moreover, spike sorting employing principal component analysis (PCA) confirms that single microchannels support the recording of multiple neurons. Overall, this work suggests a strategy to endow conventional culture plasticware with added functionality to enable cost-efficient network electrophysiology.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Electrofisiología/métodos , Neuronas/citología , Neuronas/fisiología , Neurociencias/métodos , Plásticos/química , Dióxido de Carbono/química , Costos y Análisis de Costo , Dimetilpolisiloxanos/química , Conductividad Eléctrica , Espacio Extracelular/metabolismo , Microelectrodos , Microfluídica , Técnicas de Placa-Clamp , Poliestirenos/química , Transfección
11.
Neurobiol Dis ; 30(2): 243-54, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18374587

RESUMEN

Alzheimer's disease and prion diseases (e.g., Creutzfeldt-Jakob disease) display profound neural lesions associated with aberrant protein processing and extracellular amyloid deposits. However, the intracellular events in prion diseases and their relation with the processing of the amyloid precursor protein (APP) and beta-amyloid generation are unknown. The adaptor protein Dab1 may regulate intracellular trafficking and secretase-mediated proteolysis in APP processing. However, a putative relationship between prion diseases and Dab1/APP interactions is lacking. Thus, we examined, in inoculated animals, whether Dab1 and APP processing are targets of the intracellular events triggered by extracellular exposure to PrP(106-126) peptide. Our in vitro results indicate that PrP(106-126) peptide induces tyrosine phosphorylation of Dab1 by activated members of the Src family of tyrosine kinases (SFK), which implies further Dab1 degradation. We also corroborate these results in Dab1 protein levels in prion-inoculated hamsters. Finally, we show that fibrillar prion peptides have a dual effect on APP processing and beta-amyloid production. First, they block APP trafficking at the cell membrane, thus decreasing beta-amyloid production. In parallel, they reduce Dab1 levels, which also alter APP processing. Lastly, neuronal cultures from Dab1-deficient mice showed severe impairment of APP processing with reduced sAPP secretion and A beta production after prion peptide incubation. Taken together, these data indicate a link between intracellular events induced by exposure to extracellular fibrillar peptide or PrP(res), and APP processing and implicate Dab1 in this link.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/fisiología , Priones/fisiología , Procesamiento Proteico-Postraduccional , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/biosíntesis , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Cricetinae , Femenino , Humanos , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/deficiencia , Fragmentos de Péptidos/genética , Fosforilación , Proteína PrP 27-30/farmacología , Embarazo , Priones/genética , Procesamiento Proteico-Postraduccional/fisiología
12.
Free Radic Biol Med ; 45(8): 1159-66, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18703134

RESUMEN

The basic molecular underpinnings of the pathological changes that unfold in prion disease remain elusive. A key role of increased oxidative stress has been hypothesized. Given the transient nature of most intermediate molecules implicated, increased oxidative stress is better assessed by quantitating the damage it causes to macromolecules. We used mass spectrometry-based methods to measure specific products of protein oxidation, glycoxidation, and lipoxidation in brains from patients suffering from Creutzfeldt-Jakob disease and Syrian hamsters affected by scrapie. In both cases, increased amounts of glutamic and aminoadipic semialdehydes, products of metal-catalyzed oxidation, malondialdehydelysine (a product of lipoxidation), N-epsilon-carboxyethyllysine (a product of glycoxidation), and N-epsilon-carboxymethyllysine (generated by lipoxidation and glycoxidation) were measured. PrP(Sc), the infectious isoform of the prion protein that accumulates in prion disease, was itself shown to be a target of increased oxidative modification. These changes were accompanied by alterations in fatty acid composition and increased phosphorylation of ERK(1/2) and p38, protein kinases known to respond to increased flows of ROS. These data support an important role of oxidative damage in the pathology of prion disease.


Asunto(s)
Química Encefálica , Encéfalo/metabolismo , Estrés Oxidativo/fisiología , Enfermedades por Prión/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/patología , Cricetinae , Ácidos Grasos/análisis , Ácidos Grasos/metabolismo , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Peroxidación de Lípido , Masculino , Mesocricetus , Persona de Mediana Edad , Oxidación-Reducción
13.
FASEB J ; 21(12): 3107-17, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17494993

RESUMEN

Prnp knockout mice that overexpress an amino-truncated form of PrPc (deltaPrP) are ataxic and display cerebellar cell loss and premature death. Studies on the molecular and intracellular events that trigger cell death in these mutants may contribute to elucidate the functions of PrPc and to the design of treatments for prion disease. Here we examined the effects of Bcl-2 overexpression in neurons on the development of the neurological syndrome and cerebellar pathology of deltaPrP. We show that deltaPrP overexpression activates the stress-associated kinases ERK1-2 in reactive astroglia, p38 and the phosphorylation of p53, which leads to the death of cerebellar neurons in mutant mice. We found that the expression of deltaPrP in cell lines expressing very low levels of PrPc strongly induces the activation of apoptotic pathways, thereby leading to caspase-3 activation and cell death, which can be prevented by coexpressing Bcl-2. Finally, we corroborate in vivo that neuronal-directed Bcl-2 overexpression in deltaPrP mice (deltaPrP Bcl-2) markedly reduces caspase-3 activation, glial activation, and neuronal cell death in cerebellum by improving locomotor deficits and life expectancy.


Asunto(s)
Caspasa 3/metabolismo , Enfermedades Cerebelosas/patología , Proteínas PrPC , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Conducta Animal/fisiología , Caspasa 3/genética , Muerte Celular , Células Cultivadas , Enfermedades Cerebelosas/metabolismo , Cerebelo/citología , Cerebelo/metabolismo , Cerebelo/patología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas/citología , Neuronas/fisiología , Fenotipo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Prog Neurobiol ; 165-167: 87-102, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29530723

RESUMEN

Several studies have indicated that certain misfolded amyloids composed of tau, ß-amyloid or α-synuclein can be transferred from cell to cell, suggesting the contribution of mechanisms reminiscent of those by which infective prions spread through the brain. This process of a 'prion-like' spreading between cells is also relevant as a novel putative therapeutic target that could block the spreading of proteinaceous aggregates throughout the brain which may underlie the progressive nature of neurodegenerative diseases. The relevance of ß-amyloid oligomers and cellular prion protein (PrPC) binding has been a focus of interest in Alzheimer's disease (AD). At the molecular level, ß-amyloid/PrPC interaction takes place in two differently charged clusters of PrPC. In addition to ß-amyloid, participation of PrPC in α-synuclein binding and brain spreading also appears to be relevant in α-synucleopathies. This review summarizes current knowledge about PrPC as a putative receptor for amyloid proteins and the physiological consequences of these interactions.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Interneuronas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas Priónicas/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Humanos
15.
Mol Neurobiol ; 55(3): 1847-1860, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28229331

RESUMEN

The cellular prion protein, encoded by the gene Prnp, has been reported to be a receptor of ß-amyloid. Their interaction is mandatory for neurotoxic effects of ß-amyloid oligomers. In this study, we aimed to explore whether the cellular prion protein participates in the spreading of α-synuclein. Results demonstrate that Prnp expression is not mandatory for α-synuclein spreading. However, although the pathological spreading of α-synuclein can take place in the absence of Prnp, α-synuclein expanded faster in PrPC-overexpressing mice. In addition, α-synuclein binds strongly on PrPC-expressing cells, suggesting a role in modulating the effect of α-synuclein fibrils.


Asunto(s)
Neuronas/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Transporte de Proteínas/fisiología
16.
Mol Neurobiol ; 55(4): 3033-3048, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28466265

RESUMEN

Gerstmann-Sträussler-Scheinker (GSS) syndrome is a fatal autosomal dominant neurodegenerative prionopathy clinically characterized by ataxia, spastic paraparesis, extrapyramidal signs and dementia. In some GSS familiar cases carrying point mutations in the PRNP gene, patients also showed comorbid tauopathy leading to mixed pathologies. In this study we developed an induced pluripotent stem (iPS) cell model derived from fibroblasts of a GSS patient harboring the Y218N PRNP mutation, as well as an age-matched healthy control. This particular PRNP mutation is unique with very few described cases. One of the cases presented neurofibrillary degeneration with relevant Tau hyperphosphorylation. Y218N iPS-derived cultures showed relevant astrogliosis, increased phospho-Tau, altered microtubule-associated transport and cell death. However, they failed to generate proteinase K-resistant prion. In this study we set out to test, for the first time, whether iPS cell-derived neurons could be used to investigate the appearance of disease-related phenotypes (i.e, tauopathy) identified in the GSS patient.


Asunto(s)
Enfermedad de Gerstmann-Straussler-Scheinker/genética , Enfermedad de Gerstmann-Straussler-Scheinker/patología , Células Madre Pluripotentes Inducidas/patología , Mutación/genética , Proteínas Priónicas/genética , Proteínas tau/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Secuencia de Bases , Encéfalo/patología , Diferenciación Celular , Células Cultivadas , Femenino , Gliosis/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Persona de Mediana Edad , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación
17.
Prion ; 11(4): 226-233, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28759332

RESUMEN

The term 'prion-like' is used to define some misfolded protein species that propagate intercellularly, triggering protein aggregation in recipient cells. For cell binding, both direct plasma membrane interaction and membrane receptors have been described for particular amyloids. In this respect, emerging evidence demonstrates that several ß-sheet enriched proteins can bind to the cellular prion protein (PrPC). Among other interactions, the physiological relevance of the binding between ß-amyloid and PrPC has been a relevant focus of numerous studies. At the molecular level, published data point to the second charged cluster domain of the PrPC molecule as the relevant binding domain of the ß-amyloid/PrPC interaction. In addition to ß-amyloid, participation of PrPC in binding α-synuclein, responsible for neurodegenerative synucleopathies, has been reported. Although results indicate relevant participation of PrPC in the spreading of α-synuclein in living mice, the physiological relevance of the interaction remains elusive. In this comment, we focus our attention on summarizing current knowledge of PrPC as a receptor for amyloid proteins and its physiological significance, with particular focus on α-synuclein.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas PrPC/metabolismo , Células Receptoras Sensoriales/metabolismo , alfa-Sinucleína/metabolismo , Animales , Ratones
18.
Mol Neurobiol ; 54(8): 6412-6425, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27726110

RESUMEN

Reelin is an extracellular glycoprotein involved in key cellular processes in developing and adult nervous system, including regulation of neuronal migration, synapse formation, and plasticity. Most of these roles are mediated by the intracellular phosphorylation of disabled-1 (Dab1), an intracellular adaptor molecule, in turn mediated by binding Reelin to its receptors. Altered expression and glycosylation patterns of Reelin in cerebrospinal and cortical extracts have been reported in Alzheimer's disease. However, putative changes in Reelin are not described in natural prionopathies or experimental models of prion infection or toxicity. With this is mind, in the present study, we determined that Reelin protein and mRNA levels increased in CJD human samples and in mouse models of human prion disease in contrast to murine models of prion infection. However, changes in Reelin expression appeared only at late terminal stages of the disease, which prevent their use as an efficient diagnostic biomarker. In addition, increased Reelin in CJD and in in vitro models does not correlate with Dab1 phosphorylation, indicating failure in its intracellular signaling. Overall, these findings widen our understanding of the putative changes of Reelin in neurodegeneration.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Síndrome de Creutzfeldt-Jakob/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades por Prión/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/metabolismo , Síndrome de Creutzfeldt-Jakob/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Fosforilación , Enfermedades por Prión/genética , Proteína Reelina
19.
Prion ; 10(1): 25-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890218

RESUMEN

Since its discovery the cellular prion protein (encoded by the Prnp gene) has been associated with a large number of functions. The proposed functions rank from basic cellular processes such as cell cycle and survival to neural functions such as behavior and neuroprotection, following a pattern similar to that of Moore's law for electronics. In addition, particular interest is increasing in the participation of Prnp in neurodegeneration. However, in recent years a redefinition of these functions has begun, since examples of previously attributed functions were increasingly re-associated with other proteins. Most of these functions are linked to so-called "Prnp-flanking genes" that are close to the genomic locus of Prnp and which are present in the genome of some Prnp mouse models. In addition, their role in neuroprotection against convulsive insults has been confirmed in recent studies. Lastly, in recent years a large number of models indicating the participation of different domains of the protein in apoptosis have been uncovered. However, after more than 10 years of molecular dissection our view is that the simplest mechanistic model in PrP(C)-mediated cell death should be considered, as Ockham's razor theory suggested.


Asunto(s)
Proteínas Priónicas , Animales , Ratones , Ratones Noqueados , Plasticidad Neuronal , Enfermedades por Prión , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Proteínas Priónicas/fisiología
20.
Mol Neurobiol ; 53(7): 4438-48, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26250617

RESUMEN

The biological functions of the cellular prion protein remain poorly understood. In fact, numerous studies have aimed to determine specific functions for the different protein domains. Studies of cellular prion protein (PrP(C)) domains through in vivo expression of molecules carrying internal deletions in a mouse Prnp null background have provided helpful data on the implication of the protein in signalling cascades in affected neurons. Nevertheless, understanding of the mechanisms underlying the neurotoxicity induced by these PrP(C) deleted forms is far from complete. To better define the neurotoxic or neuroprotective potential of PrP(C) N-terminal domains, and to overcome the heterogeneity of results due to the lack of a standardized model, we used neuroblastoma cells to analyse the effects of overexpressing PrP(C) deleted forms. Results indicate that PrP(C) N-terminal deleted forms were properly processed through the secretory pathway. However, PrPΔF35 and PrPΔCD mutants led to death by different mechanisms sharing loss of alpha-cleavage and activation of caspase-3. Our data suggest that both gain-of-function and loss-of-function pathogenic mechanisms may be associated with N-terminal domains and may therefore contribute to neurotoxicity in prion disease. Dissecting the molecular response induced by PrPΔF35 may be the key to unravelling the physiological and pathological functions of the prion protein.


Asunto(s)
Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Neuroblastoma/metabolismo , Proteínas Priónicas/metabolismo , Animales , Muerte Celular , Línea Celular Tumoral , Microdominios de Membrana/metabolismo , Ratones , Modelos Biológicos , Neuroblastoma/patología , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA