Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Intervalo de año de publicación
1.
NPJ Parkinsons Dis ; 10(1): 116, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879633

RESUMEN

Most neurodegenerative diseases lack definitive diagnostic tests, and the identification of easily accessible and reliable biomarkers remains a critical unmet need. Since tau protein is highly expressed in skin of tauopathies patients, we aimed to exploit the ultrasensitive seeding activity assay (SAA) to assess tau seeding activity in skin of patients with tauopathies. In this multicentric, case-control study, patients with tauopathies and synucleinopathies were consecutively recruited and sex-matched to healthy controls (HC). Subjects underwent a double 3 mm skin biopsy in cervical area and ankle. Skin tau-SAA, using TauK18 and TauK19 as reaction substrates for 4R and 3R isoforms, seeding score, clinical scales, biochemical and morphological characterization of SAA end-products were evaluated. We analyzed 58 subjects: 24 tauopathies (18 progressive supranuclear palsy, PSP, and 6 corticobasal degeneration, CBD), 20 synucleinopathies (14 Parkinson's disease, PD, and 6 multiple system atrophy, MSA), and 14 HC. PSP and CBD showed higher tau seeding activity at both anatomical sites. A greater sensitivity of 4R-SAA than 3R-SAA was observed. 4R tau-SAA identified tauopathies with 71% sensitivity and 93% specificity. Accuracy was higher for PSP than CBD: PSP vs HC / PD (AUC 0.825), while CBD vs HC / PD (AUC 0.797), and PSP vs MSA (AU 0.778). SAA end-products showed differences in biochemical and morphological characterization according to the anatomical site. Skin tau-SAA identifies tauopathies with good accuracy and can be used to implement the in-vivo clinical diagnosis of patients with neurodegenerative diseases. Further characterization of peripheral tau seed in skin may elucidate the structure of tau deposits in brain.

2.
Cell Death Dis ; 13(12): 1048, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36526614

RESUMEN

Parkinson's disease (PD) presents the selective loss of A9 dopaminergic (DA) neurons of Substantia Nigra pars compacta (SNpc) and the presence of intracellular aggregates called Lewy bodies. α-synuclein (α-syn) species truncated at the carboxy-terminal (C-terminal) accumulate in pathological inclusions and promote α-syn aggregation and toxicity. Haemoglobin (Hb) is the major oxygen carrier protein in erythrocytes. In addition, Hb is expressed in A9 DA neurons where it influences mitochondrial activity. Hb overexpression increases cells' vulnerability in a neurochemical model of PD in vitro and forms cytoplasmic and nucleolar aggregates upon short-term overexpression in mouse SNpc. In this study, α and ß-globin chains were co-expressed in DA cells of SNpc in vivo upon stereotaxic injections of an Adeno-Associated Virus isotype 9 (AAV9) and in DA iMN9D cells in vitro. Long-term Hb over-expression in SNpc induced the loss of about 50% of DA neurons, mild motor impairments, and deficits in recognition and spatial working memory. Hb triggered the formation of endogenous α-syn C-terminal truncated species. Similar α-syn fragments were found in vitro in DA iMN9D cells over-expressing α and ß- globins when treated with pre-formed α-syn fibrils. Our study positions Hb as a relevant player in PD pathogenesis for its ability to trigger DA cells' loss in vivo and the formation of C-terminal α-syn fragments.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Ratones , Animales , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neuronas Dopaminérgicas/metabolismo , Sustancia Negra/metabolismo , Enfermedad de Parkinson/metabolismo , Hemoglobinas/metabolismo , Cognición
3.
Mol Neurobiol ; 59(3): 1577-1588, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35000151

RESUMEN

The cellular prion protein (PrPC) is a ubiquitous glycoprotein highly expressed in the brain where it is involved in neurite outgrowth, copper homeostasis, NMDA receptor regulation, cell adhesion, and cell signaling. Conformational conversion of PrPC into its insoluble and aggregation-prone scrapie form (PrPSc) is the trigger for several rare devastating neurodegenerative disorders, collectively referred to as prion diseases. Recent work indicates that the ubiquitin-proteasome system is involved in quality control of PrPC. To better dissect the role of ubiquitination in PrPC physiology, we focused on the E3 RING ubiquitin ligase tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6). Here, we report that PrPC interacts with TRAF6 both in vitro, in cells, and in vivo, in the mouse brain. Transient overexpression of TRAF6 indirectly modulates PrPC ubiquitination and triggers redistribution of PrPC into the insoluble fraction. Importantly, in the presence of wild-type TRAF6, but not a mutant lacking E3 ligase activity, PrPC accumulates into cytoplasmic aggresome-like inclusions containing TRAF6 and p62/SQSTM1. Our results suggest that TRAF6 ligase activity could exert a role in the regulation of PrPC redistribution in cells under physiological conditions. This novel interaction may uncover possible mechanisms of cell clearance/reorganization in prion diseases.


Asunto(s)
Factor 6 Asociado a Receptor de TNF , Ubiquitina-Proteína Ligasas , Animales , Ratones , Proteínas Priónicas/metabolismo , Unión Proteica , Proteína Sequestosoma-1/metabolismo , Solubilidad , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
4.
Viruses ; 13(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34452489

RESUMEN

Cytoplasmic aggregation of the primarily nuclear TAR DNA-binding protein 43 (TDP-43) affects neurons in most amyotrophic lateral sclerosis (ALS) and approximately half of frontotemporal lobar degeneration (FTLD) cases. The cellular prion protein, PrPC, has been recognized as a common receptor and downstream effector of circulating neurotoxic species of several proteins involved in neurodegeneration. Here, capitalizing on our recently adapted TDP-43 real time quaking induced reaction, we set reproducible protocols to obtain standardized preparations of recombinant TDP-43 fibrils. We then exploited two different cellular systems (human SH-SY5Y and mouse N2a neuroblastoma cells) engineered to express low or high PrPC levels to investigate the link between PrPC expression on the cell surface and the internalization of TDP-43 fibrils. Fibril uptake was increased in cells overexpressing either human or mouse prion protein. Increased internalization was associated with detrimental consequences in all PrP-overexpressing cell lines but was milder in cells expressing the human form of the prion protein. As described for other amyloids, treatment with TDP-43 fibrils induced a reduction in the accumulation of the misfolded form of PrPC, PrPSc, in cells chronically infected with prions. Our results expand the list of misfolded proteins whose uptake and detrimental effects are mediated by PrPC, which encompass almost all pathological amyloids involved in neurodegeneration.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas Priónicas/metabolismo , Animales , Transporte Biológico , Línea Celular , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/farmacología , Humanos , Ratones , Proteínas PrPC/clasificación , Proteínas Priónicas/genética
5.
J Extracell Vesicles ; 10(9): e12114, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34276899

RESUMEN

Astrocytes-derived extracellular vesicles (EVs) are key players in glia-neuron communication. However, whether EVs interact with neurons at preferential sites and how EVs reach these sites on neurons remains elusive. Using optical manipulation to study single EV-neuron dynamics, we here show that large EVs scan the neuron surface and use neuronal processes as highways to move extracellularly. Large EV motion on neurites is driven by the binding of EV to a surface receptor that slides on neuronal membrane, thanks to actin cytoskeleton rearrangements. The use of prion protein (PrP)-coated synthetic beads and PrP knock out EVs/neurons points at vesicular PrP and its receptor(s) on neurons in the control of EV motion. Surprisingly, a fraction of large EVs contains actin filaments and has an independent capacity to move in an actin-mediated way, through intermittent contacts with the plasma membrane. Our results unveil, for the first time, a dual mechanism exploited by astrocytic large EVs to passively/actively reach target sites on neurons moving on the neuron surface.


Asunto(s)
Astrocitos/citología , Vesículas Extracelulares/fisiología , Neuritas/fisiología , Proteínas Priónicas/metabolismo , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Citoesqueleto/fisiología , Metabolismo Energético , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Propiedades de Superficie
6.
Cells ; 11(1)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35011649

RESUMEN

Parkinson's disease (PD) and multiple system atrophy (MSA) are caused by two distinct strains of disease-associated α-synuclein (αSynD). Recently, we have shown that olfactory mucosa (OM) samples of patients with PD and MSA can seed the aggregation of recombinant α-synuclein by means of Real-Time Quaking-Induced Conversion (αSyn_RT-QuIC). Remarkably, the biochemical and morphological properties of the final α-synuclein aggregates significantly differed between PD and MSA seeded samples. Here, these aggregates were given to neuron-like differentiated SH-SY5Y cells and distinct inflammatory responses were observed. To deepen whether the morphological features of α-synuclein aggregates were responsible for this variable SH-SY5Y inflammatory response, we generated three biochemically and morphologically distinct α-synuclein aggregates starting from recombinant α-synuclein that were used to seed αSyn_RT-QuIC reaction; the final reaction products were used to stimulate SH-SY5Y cells. Our study showed that, in contrast to OM samples of PD and MSA patients, the artificial aggregates did not transfer their distinctive features to the αSyn_RT-QuIC products and the latter induced analogous inflammatory responses in cells. Thus, the natural composition of the αSynD strains but also other specific factors in OM tissue can substantially modulate the biochemical, morphological and inflammatory features of the αSyn_RT-QuIC products.


Asunto(s)
Inflamación/patología , Atrofia de Múltiples Sistemas/metabolismo , Atrofia de Múltiples Sistemas/patología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Neuroblastoma/patología , Agregado de Proteínas , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/ultraestructura
7.
J Neurochem ; 155(5): 577-591, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32394432

RESUMEN

Tauopathies are prevalent, invariably fatal brain diseases for which no cure is available. Tauopathies progressively affect the brain through cell-to-cell transfer of tau protein amyloids, yet the spreading mechanisms remain unknown. Here we show that the cellular prion protein (PrPC ) facilitates the uptake of tau aggregates by cultured cells, possibly by acting as an endocytic receptor. In mouse neuroblastoma cells, pull-down experiments revealed that tau amyloids bind to PrPC . Confocal images of both wild-type and PrPC -knockout N2a cells treated with fluorescently labeled synthetic tau fibrils showed that the internalization was reduced in isogenic cells devoid of the gene encoding PrPC . Pre-treatment of the same cells with antibodies against N-proximal epitopes of PrPC impaired the binding of tau amyloids and decreased their uptake. Surprisingly, exposure of chronically prion-infected cells to tau amyloids reduced the accumulation of aggregated prion protein and this effect lasted for more than 72 hr after amyloid removal. These results point to bidirectional interactions between the two proteins: while PrPC mediates the entrance of tau fibrils in cells, PrPSc buildup is greatly reduced in their presence, possibly because of an impairment in the prion conversion process.


Asunto(s)
Amiloide/metabolismo , Proteínas PrPC/metabolismo , Proteínas tau/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Ratones , Proteínas Priónicas/metabolismo , Unión Proteica/fisiología
8.
Eur J Med Chem ; 196: 112295, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32325366

RESUMEN

Prion diseases or transmissible spongiform encephalopathies (TSEs) are a group of rare neurodegenerative disorders. TSEs are characterized by the accumulation of prions (PrPSc) that represent pathological isoforms of the physiological cellular prion protein PrPC. Although the conversion of PrPC to PrPSc is still not completely understood, blocking this process may lead to develop new therapies. Here, we have generated a pharmacophore model, based on anti-prion molecules reported in literature to be effective in phenotypic assay. The model was used to conduct a virtual screen of commercial compound databases that selected a small library of ten compounds. These molecules were then screened in mouse neuroblastoma cell line chronically infected with prions (ScN2a) after excluding neurotoxicity. 1 has been identified as the therapeutic hit on the basis of the following evidence: chronic treatments of ScN2a cells using 1 eliminate PrPSc loaded in both Western blotting analysis and Real-Time Quaking-Induced Conversion (RT-QuIC) assay. We also proposed the mechanism of action of 1 by which it has the ability to bind PrPC and consequentially blocks prion conversion. Herein we describe the results of these efforts.


Asunto(s)
Fenotiazinas/farmacología , Proteínas Priónicas/antagonistas & inhibidores , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Ratones , Modelos Moleculares , Estructura Molecular , Fenotiazinas/química , Proteínas Priónicas/aislamiento & purificación , Proteínas Priónicas/metabolismo , Relación Estructura-Actividad Cuantitativa
9.
Int J Biol Macromol ; 154: 206-216, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32179119

RESUMEN

The notion that nanoscale surfaces influence protein conformational transitions stimulates the investigation of fibrillogenic polypeptides adsorbing to nanomaterials. Alpha-synuclein (αS) is a prototypical amyloidogenic protein whose aggregation is associated with severe neurodegenerative disorders. We explored the interaction of αS with silica nanoparticles (SNPs) in diverse solution conditions, ranging from protein-free to protein-rich media. We found that the SNP-binding region of αS, determined by site-resolved NMR spectroscopy, was similar in simple buffer and blood serum. Competition binding experiments with isotopic homologues and different proteins showed that cosolutes elicited molecular exchange in a protein-specific manner. The interaction of an oxidized, fibrillation-resistant protein form with SNPs was similar to that of unmodified αS. SNPs, however, did not stimulate fibrillation of the oxidized protein, which remained fibrillation incompetent. CD experiments revealed SNP-induced perturbations of the structural properties of oxidized and non-oxidized αS. Thus, while αS binding to SNPs is essentially orthogonal to fibril formation, the interaction perturbs the distribution of conformational states populated by the protein in the colloidal suspension. This study sheds light on the dynamic nature of αS interactions with NPs, an aspect that crucially impacts on our ability to control aggregation of αS.


Asunto(s)
Nanopartículas/química , Agregación Patológica de Proteínas , Proteínas Recombinantes/química , Dióxido de Silicio/química , alfa-Sinucleína/química , Humanos , Unión Proteica , Conformación Proteica , Pliegue de Proteína
10.
Nanoscale ; 12(14): 7631-7640, 2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32104855

RESUMEN

The aberrant misfolding and aggregation of alpha synuclein (αS) into toxic oligomeric species is one of the key features associated with the pathogenesis of Parkinson's disease (PD). It involves different biochemical and biophysical factors as plasma membrane binding and interaction with heavy metal ions. In the present work, atomic force microscopy (AFM) is combined with Fourier Transform Infrared Spectroscopy (FTIR) measurements to investigate the interaction of wild-type (WT) and A53T mutated alpha synuclein with artificial lipid bilayers mimicking lipid raft (LR) domains, before and after ferrous cations (Fe2+) treatment. In the absence of iron, protein monomers produce a thinning of the membrane, targeting the non-raft phase of the bilayer preferentially. On the contrary, iron actively promotes the formation of globular protein aggregates, resembling oligomers, targeted to LR domains. In both aggregation states, monomer and oligomer, the mutated A53T protein exhibits a greater and faster membrane-interaction. These results underlie a new mechanism of membrane-protein interaction in PD. The targeting of Fe2+-promoted αS oligomers to LRs might be functional for the disease and be helpful for the development of new therapeutic strategies.


Asunto(s)
Hierro/química , Microdominios de Membrana/química , alfa-Sinucleína/química , Compuestos Ferrosos/química , Compuestos Ferrosos/metabolismo , Humanos , Hierro/metabolismo , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/metabolismo , Microscopía de Fuerza Atómica , Mutagénesis Sitio-Dirigida , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Agregado de Proteínas , Unión Proteica , Espectroscopía Infrarroja por Transformada de Fourier , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
11.
J Med Chem ; 61(17): 7640-7656, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30078314

RESUMEN

Several findings propose the altered tau protein network as an important target for Alzheimer's disease (AD). Particularly, two points of pharmacological intervention can be envisaged: inhibition of phosphorylating tau kinase GSK-3ß and tau aggregation process. On the basis of this consideration and on our interest in multitarget paradigms in AD, we report on the discovery of 2,4-thiazolidinedione derivatives endowed with such a profile. 28 and 30 displayed micromolar IC50 values toward GSK-3ß, together with the capacity of inhibiting AcPHF6 aggregation of 60% and 80% at 10 µM, respectively. In addition, they showed PAMPA-BBB permeability, together with a suitable cellular safety profile. 30 also displayed inhibition of both K18 and full-length tau aggregations. Finally, both compounds were able to improve cell viability in an okadaic acid-induced neurodegeneration cell model. To the best of our knowledge, 28 and 30 are the first balanced, nontoxic, dual-acting compounds hitting tau cascade at two different hubs.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas tau/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Fármacos del Sistema Nervioso Central/efectos adversos , Fármacos del Sistema Nervioso Central/química , Fármacos del Sistema Nervioso Central/farmacología , Dicroismo Circular , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Transferencia Resonante de Energía de Fluorescencia , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Células Hep G2 , Humanos , Microscopía de Fuerza Atómica , Terapia Molecular Dirigida/métodos , Ácido Ocadaico/toxicidad , Fosforilación/efectos de los fármacos , Ratas , Relación Estructura-Actividad , Porcinos , Tiazolidinedionas/química , Proteínas tau/antagonistas & inhibidores
12.
Sci Rep ; 7(1): 10050, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855681

RESUMEN

The precise molecular mechanism of how misfolded α-synuclein (α-Syn) accumulates and spreads in synucleinopathies is still unknown. Here, we show the role of the cellular prion protein (PrPC) in mediating the uptake and the spread of recombinant α-Syn amyloids. The in vitro data revealed that the presence of PrPC fosters the higher uptake of α-Syn amyloid fibrils, which was also confirmed in vivo in wild type (Prnp +/+) compared to PrP knock-out (Prnp -/-) mice. Additionally, the presence of α-Syn amyloids blocked the replication of scrapie prions (PrPSc) in vitro and ex vivo, indicating a link between the two proteins. Indeed, whilst PrPC is mediating the internalization of α-Syn amyloids, PrPSc is not able to replicate in their presence. This observation has pathological relevance, since several reported case studies show that the accumulation of α-Syn amyloid deposits in Creutzfeldt-Jakob disease patients is accompanied by a longer disease course.


Asunto(s)
Amiloide/metabolismo , Encéfalo/patología , Síndrome de Creutzfeldt-Jakob/metabolismo , Neuronas/metabolismo , Proteínas Priónicas/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/administración & dosificación , Amiloide/genética , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/patología , Endopeptidasa K/química , Regulación de la Expresión Génica , Humanos , Inyecciones Intraventriculares , Ratones , Ratones Noqueados , Neuronas/patología , Proteínas Priónicas/genética , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Técnicas Estereotáxicas , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/genética
13.
Eur J Med Chem ; 127: 859-873, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27842893

RESUMEN

Prion diseases are serious, not curable neurodegenerative disorders caused by the accumulation of the misfolded protein PrPSc that represents the pathological variant of the normally folded cellular protein PrPC. Molecules that bind the cellular isoform PrPC preventing its misfolding, could arrest the progression of pathological conditions related to the abnormal PrP protein. In this context, by combining 3D-QSAR model, derived from pharmacophore-based alignment, with molecular docking procedures and physico-chemical properties prediction we have developed a virtual screening protocol to find novel chemicals able to prevent PrPC misfolding. We identified different hits characterized by low toxicity and able to inhibit PrPSc accumulation in vitro in prion-infected neuroblastoma cell lines (ScN2a). In this assay, the pyrroloquinoxaline hydrazone 96 showed the higest potency with an IC50 value of 1.6 µM. Pyrroloquinoxaline 96 was demonstrated also to bind PrPSc aggregates in infected ScN2a cells with a fluorescence pattern comparable to that found for Thioflavin-T. In consideration of its satisfactory physico-chemical properties, including predicted blood brain barrier permeability, 96 could represent an interesting prototypic hit for the development of diagnostic and therapeutic probes for prion diseases.


Asunto(s)
Colorantes Fluorescentes/farmacología , Proteínas PrPSc/química , Enfermedades por Prión/metabolismo , Agregado de Proteínas/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Proteínas PrPSc/metabolismo , Conformación Proteica , Relación Estructura-Actividad Cuantitativa
14.
Front Neurosci ; 10: 437, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27729845

RESUMEN

Essential elements as copper and iron modulate a wide range of physiological functions. Their metabolism is strictly regulated by cellular pathways, since dysregulation of metal homeostasis is responsible for many detrimental effects. Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and prion diseases are characterized by alterations of metal ions. These neurodegenerative maladies involve proteins that bind metals and mediate their metabolism through not well-defined mechanisms. Prion protein, for instance, interacts with divalent cations via multiple metal-binding sites and it modulates several metal-dependent physiological functions, such as S-nitrosylation of NMDA receptors. In this work we focused on the effect of prion protein absence on copper and iron metabolism during development and adulthood. In particular, we investigated copper and iron functional values in serum and several organs such as liver, spleen, total brain and isolated hippocampus. Our results show that iron content is diminished in prion protein-null mouse serum, while it accumulates in liver and spleen. Our data suggest that these alterations can be due to impairments in copper-dependent cerulopalsmin activity which is known to affect iron mobilization. In prion protein-null mouse total brain and hippocampus, metal ion content shows a fluctuating trend, suggesting the presence of homeostatic compensatory mechanisms. However, copper and iron functional values are likely altered also in these two organs, as indicated by the modulation of metal-binding protein expression levels. Altogether, these results reveal that the absence of the cellular prion protein impairs copper metabolism and copper-dependent oxidase activity, with ensuing alteration of iron mobilization from cellular storage compartments.

15.
Sci Rep ; 5: 15253, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26482532

RESUMEN

The conversion of the prion protein (PrP(C)) into prions plays a key role in transmissible spongiform encephalopathies. Despite the importance for pathogenesis, the mechanism of prion formation has escaped detailed characterization due to the insoluble nature of prions. PrP(C) interacts with copper through octarepeat and non-octarepeat binding sites. Copper coordination to the non-octarepeat region has garnered interest due to the possibility that this interaction may impact prion conversion. We used X-ray absorption spectroscopy to study copper coordination at pH 5.5 and 7.0 in human PrP(C) constructs, either wild-type (WT) or carrying pathological mutations. We show that mutations and pH cause modifications of copper coordination in the non-octarepeat region. In the WT at pH 5.5, copper is anchored to His96 and His111, while at pH 7 it is coordinated by His111. Pathological point mutations alter the copper coordination at acidic conditions where the metal is anchored to His111. By using in vitro approaches, cell-based and computational techniques, we propose a model whereby PrP(C) coordinating copper with one His in the non-octarepeat region converts to prions at acidic condition. Thus, the non-octarepeat region may act as the long-sought-after prion switch, critical for disease onset and propagation.


Asunto(s)
Sitios de Unión , Cobre/química , Priones/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular Tumoral , Secuencia Conservada , Cobre/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Priones/genética , Priones/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Secuencias Repetitivas de Ácidos Nucleicos , Relación Estructura-Actividad
16.
Expert Opin Drug Discov ; 10(4): 389-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25682812

RESUMEN

INTRODUCTION: Recent years have witnessed major advances in our understanding of the molecular bases of prion diseases. These studies not only highlight the protein misfolding as a potential initiator of a neurodegenerative process, they also provide a foundation for considering whether such a process can be common to many neurodegenerative diseases, including Alzheimer's disease. This makes prion diseases a sort of prototype of neurodegenerative disease, endowed with some intrinsic positive features in terms of drug development. Thanks to the fact that disappearance of the scrapie protein can serve as a clear readout of drug efficiency, phenotypic approaches have high potential for prion disease drug discovery. AREAS COVERED: In this review, the authors discuss phenotypic screening and how it lends itself to drug repositioning. Furthermore, they discuss the advantages of working with a molecule with proven safety, tolerability and drug-like properties in combination with a reliable phenotypic screening and how it could improve the success rate for prion drug development. They also provide examples of several interesting candidates that have been identified using this approach, including quinacrine, astemizole, guanabenz and doxycycline. EXPERT OPINION: The availability of persistently scrapie-infected murine neuroblastoma cells has greatly helped to identify compounds that inhibit prion formation. However, a human neuronal model infected with the human isoform would ultimately serve as the ideal disease model toward the discovery of effective drugs.


Asunto(s)
Diseño de Fármacos , Enfermedades por Prión/tratamiento farmacológico , Priones/metabolismo , Animales , Descubrimiento de Drogas/métodos , Humanos , Ratones , Fenotipo , Pliegue de Proteína
17.
PLoS Pathog ; 11(12): e1005354, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26720726

RESUMEN

Prions are infectious proteins that possess multiple self-propagating structures. The information for strains and structural specific barriers appears to be contained exclusively in the folding of the pathological isoform, PrP(Sc). Many recent studies determined that de novo prion strains could be generated in vitro from the structural conversion of recombinant (rec) prion protein (PrP) into amyloidal structures. Our aim was to elucidate the conformational diversity of pathological recPrP amyloids and their biological activities, as well as to gain novel insights in characterizing molecular events involved in mammalian prion conversion and propagation. To this end we generated infectious materials that possess different conformational structures. Our methodology for the prion conversion of recPrP required only purified rec full-length mouse (Mo) PrP and common chemicals. Neither infected brain extracts nor amplified PrP(Sc) were used. Following two different in vitro protocols recMoPrP converted to amyloid fibrils without any seeding factor. Mouse hypothalamic GT1 and neuroblastoma N2a cell lines were infected with these amyloid preparations as fast screening methodology to characterize the infectious materials. Remarkably, a large number of amyloid preparations were able to induce the conformational change of endogenous PrPC to harbor several distinctive proteinase-resistant PrP forms. One such preparation was characterized in vivo habouring a synthetic prion with novel strain specified neuropathological and biochemical properties.


Asunto(s)
Enfermedades por Prión/patología , Priones/química , Priones/metabolismo , Secuencia de Aminoácidos , Proteínas Amiloidogénicas/química , Animales , Western Blotting , Línea Celular , Modelos Animales de Enfermedad , Ratones , Microscopía de Fuerza Atómica , Datos de Secuencia Molecular , Proteínas Priónicas , Priones/síntesis química , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química
18.
Antioxid Redox Signal ; 22(9): 772-84, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25490055

RESUMEN

AIMS: Several neurodegenerative disorders show alterations in glutamatergic synapses and increased susceptibility to excitotoxicity. Mounting evidence suggests a central role for the cellular prion protein (PrP(C)) in neuroprotection. Therefore, the loss of PrP(C) function occurring in prion disorders may contribute to the disease progression and neurodegeneration. Indeed, PrP(C) modulates N-methyl-d-aspartate receptors (NMDAR), thus preventing cell death. In this study, we show that PrP(C) and copper cooperatively inhibit NMDAR through S-nitrosylation, a post-translational modification resulting from the chemical reaction of nitric oxide (NO) with cysteines. RESULTS: Comparing wild-type Prnp (Prnp(+/+)) and PrP(C) knockout (Prnp(0/0)) mouse hippocampi, we found that GluN1 and GluN2A S-nitrosylation decrease in Prnp(0/0). Using organotypic hippocampal cultures, we found that copper chelation decreases NMDAR S-nitrosylation in Prnp(+/+) but not in Prnp(0/0). This suggests that PrP(C) requires copper to support the chemical reaction between NO and thiols. We explored PrP(C)-Cu neuroprotective role by evaluating neuron susceptibility to excitotoxicity in Prnp(+/+) and Prnp(0/0) cultures. We found that (i) PrP(C)-Cu modulates GluN2A-containing NMDAR, those inhibited by S-nitrosylation; (ii) PrP(C) and copper are interdependent to protect neurons from insults; (iii) neuronal NO synthase inhibition affects susceptibility in wild-type but not in Prnp(0/0), while (iv) the addition of a NO donor enhances Prnp(0/0) neurons survival. INNOVATION AND CONCLUSIONS: Our results show that PrP(C) and copper support NMDAR S-nitrosylation and cooperatively exert neuroprotection. In addition to NMDAR, PrP(C) may also favor the S-nitrosylation of other proteins. Therefore, this mechanism may be investigated in the context of the different cellular processes in which PrP(C) is involved.


Asunto(s)
Cobre/metabolismo , Cisteína/metabolismo , Óxido Nítrico/metabolismo , Priones/genética , Priones/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Hipocampo/citología , Ratones , Ratones Transgénicos , Neuronas/metabolismo
19.
BMC Neurosci ; 15: 69, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24898419

RESUMEN

BACKGROUND: α-Synuclein (α-syn) plays a central role in the pathogenesis of synucleinopathies, a group of neurodegenerative disorders that includes Parkinson disease, dementia with Lewy bodies and multiple system atrophy. Several findings from cell culture and mouse experiments suggest intercellular α-syn transfer. RESULTS: Through a methodology used to obtain synthetic mammalian prions, we tested whether recombinant human α-syn amyloids can promote prion-like accumulation in neuronal cell lines in vitro. A single exposure to amyloid fibrils of human α-syn was sufficient to induce aggregation of endogenous α-syn in human neuroblastoma SH-SY5Y cells. Remarkably, endogenous wild-type α-syn was sufficient for the formation of these aggregates, and overexpression of the protein was not required. CONCLUSIONS: Our results provide compelling evidence that endogenous α-syn can accumulate in cell culture after a single exposure to exogenous α-syn short amyloid fibrils. Importantly, using α-syn short amyloid fibrils as seed, endogenous α-syn aggregates and accumulates over several passages in cell culture, providing an excellent tool for potential therapeutic screening of pathogenic α-syn aggregates.


Asunto(s)
Sustancias Macromoleculares/metabolismo , Neuronas/metabolismo , Priones/clasificación , Priones/metabolismo , alfa-Sinucleína/clasificación , alfa-Sinucleína/metabolismo , Animales , Línea Celular , Humanos , Ratones
20.
J Neurosci ; 33(7): 2973-83, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407955

RESUMEN

The cellular form of prion protein PrP(C) is highly expressed in the brain, where it can be converted into its abnormally folded isoform PrP(Sc) to cause neurodegenerative diseases. Its predominant synaptic localization suggests a crucial role in synaptic signaling. Interestingly, PrP(C) is developmentally regulated and its high expression in the immature brain could be instrumental in regulating neurogenesis and cell proliferation. Here, PrP(C)-deficient (Prnp(0/0)) mice were used to assess whether the prion protein is involved in synaptic plasticity processes in the neonatal hippocampus. To this aim, calcium transients associated with giant depolarizing potentials, a hallmark of developmental networks, were transiently paired with mossy fiber activation in such a way that the two events were coincident. While this procedure caused long-term potentiation (LTP) in wild-type (WT) animals, it caused long-term depression (LTD) in Prnp(0/0) mice. Induction of LTP was postsynaptic and required the activation of cAMP-dependent protein kinase A (PKA) signaling. The induction of LTD was presynaptic and relied on G-protein-coupled GluK1 receptor and protein lipase C. In addition, at emerging CA3-CA1 synapses in WT mice, but not in Prnp(0/0) mice, pairing Schaffer collateral stimulation with depolarization of CA1 principal cells induced LTP, known to be PKA dependent. Postsynaptic infusion of a constitutively active isoform of PKA catalytic subunit Cα into CA1 and CA3 principal cells in the hippocampus of Prnp(0/0) mice caused a persistent synaptic facilitation that was occluded by subsequent pairing. These data suggest that PrP(C) plays a crucial role in regulating via PKA synaptic plasticity in the developing hippocampus.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Hipocampo/fisiología , Plasticidad Neuronal/fisiología , Proteínas PrPC/fisiología , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , AMP Cíclico/fisiología , Interpretación Estadística de Datos , Fenómenos Electrofisiológicos , Proteínas de Unión al GTP/fisiología , Hipocampo/crecimiento & desarrollo , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Noqueados , Red Nerviosa/fisiología , Técnicas de Placa-Clamp , Células Piramidales/fisiología , Receptores de Ácido Kaínico/fisiología , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA