Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 34(2): 2359-2375, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31907995

RESUMEN

Studies in mice with ablation of Prnp, the gene that encodes the cellular prion protein (PrPC ), have led to the hypothesis that PrPC is important for peripheral nerve myelin maintenance. Here, we have used a nontransgenic animal model to put this idea to the test; namely, goats that, due to a naturally occurring nonsense mutation, lack PrPC . Teased nerve fiber preparation revealed a demyelinating pathology in goats without PrPC . Affected nerves were invaded by macrophages and T cells and displayed vacuolated fibers, shrunken axons, and onion bulbs. Peripheral nerve lipid composition was similar in young goats with or without PrPC , but markedly different between corresponding groups of adult goats, reflecting the progressive nature of the neuropathy. This is the first report of a subclinical demyelinating polyneuropathy caused by loss of PrPC function in a nontransgenic mammal.


Asunto(s)
Enfermedades Desmielinizantes/inmunología , Cabras/inmunología , Vaina de Mielina/inmunología , Polineuropatías/inmunología , Proteínas PrPC/deficiencia , Animales , Enfermedades Desmielinizantes/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Vaina de Mielina/patología , Polineuropatías/patología , Proteínas PrPC/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
2.
Vet Res ; 51(1): 1, 2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924264

RESUMEN

Prion diseases are progressive and fatal, neurodegenerative disorders described in humans and animals. According to the "protein-only" hypothesis, the normal host-encoded prion protein (PrPC) is converted into a pathological and infectious form (PrPSc) in these diseases. Transgenic knockout models have shown that PrPC is a prerequisite for the development of prion disease. In Norwegian dairy goats, a mutation (Ter) in the prion protein gene (PRNP) effectively blocks PrPC synthesis. We inoculated 12 goats (4 PRNP+/+, 4 PRNP+/Ter, and 4 PRNPTer/Ter) intracerebrally with goat scrapie prions. The mean incubation time until clinical signs of prion disease was 601 days post-inoculation (dpi) in PRNP+/+ goats and 773 dpi in PRNP+/Ter goats. PrPSc and vacuolation were similarly distributed in the central nervous system (CNS) of both groups and observed in all brain regions and segments of the spinal cord. Generally, accumulation of PrPSc was limited in peripheral organs, but all PRNP+/+ goats and 1 of 4 PRNP+/Ter goats were positive in head lymph nodes. The four PRNPTer/Ter goats remained healthy, without clinical signs of prion disease, and were euthanized 1260 dpi. As expected, no accumulation of PrPSc was observed in the CNS or peripheral tissues of this group, as assessed by immunohistochemistry, enzyme immunoassay, and real-time quaking-induced conversion. Our study shows for the first time that animals devoid of PrPC due to a natural mutation do not propagate prions and are resistant to scrapie. Clinical onset of disease is delayed in heterozygous goats expressing about 50% of PrPC levels.


Asunto(s)
Resistencia a la Enfermedad/genética , Enfermedades de las Cabras/genética , Proteínas PrPC/deficiencia , Scrapie/genética , Animales , Femenino , Cabras
3.
Sci Rep ; 9(1): 13090, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511544

RESUMEN

Endothelial-to-mesenchyme-like transition (Endo-MT) of trabecular meshwork (TM) cells is known to be associated with primary open angle glaucoma (POAG). Here, we investigated whether the prion protein (PrPC), a neuronal protein known to modulate epithelial-to-mesenchymal transition in a variety of cell types, is expressed in the TM, and plays a similar role at this site. Using a combination of primary human TM cells and human, bovine, and PrP-knock-out (PrP-/-) mouse models, we demonstrate that PrPC is expressed in the TM of all three species, including endothelial cells lining the Schlemm's canal. Silencing of PrPC in primary human TM cells induces aggregation of ß1-integrin and upregulation of α-smooth muscle actin, fibronectin, collagen 1A, vimentin, and laminin, suggestive of transition to a mesenchyme-like phenotype. Remarkably, intraocular pressure is significantly elevated in PrP-/- mice relative to wild-type controls, suggesting reduced pliability of the extracellular matrix and increased resistance to aqueous outflow in the absence of PrPC. Since PrPC is cleaved by members of the disintegrin and matrix-metalloprotease family that are increased in the aqueous humor of POAG arising from a variety of conditions, it is likely that concomitant cleavage of PrPC exaggerates and confounds the pathology by inducing Endo-MT-like changes in the TM.


Asunto(s)
Células Endoteliales/citología , Glaucoma de Ángulo Abierto/metabolismo , Glaucoma de Ángulo Abierto/patología , Mesodermo/citología , Proteínas PrPC/metabolismo , Malla Trabecular/citología , Animales , Bovinos , Células Endoteliales/patología , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Mesodermo/patología , Ratones , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Malla Trabecular/metabolismo , Malla Trabecular/patología
4.
Methods Mol Biol ; 1658: 205-216, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28861792

RESUMEN

Prion diseases or transmissible spongiform encephalopathies (TSEs) are a group of neurodegenerative diseases where the misfolding of the prion protein (PrP) is a crucial event. Based on studies in TSE-affected humans and the generation of transgenic mouse models overexpressing different mutated versions of the PrP, we conclude that both wild-type and mutated PrPs exhibit differential propensity to misfold in vivo. Here, we describe a new method in vitro to assess and quantify the PrP misfolding phenomenon in order to better understand the molecular mechanisms involved in this process.


Asunto(s)
Bioensayo , Proteínas PrPC/química , Proteínas PrPSc/química , Sonicación/métodos , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Química Encefálica , Diálisis , Endopeptidasa K/química , Expresión Génica , Ratones , Ratones Noqueados , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Conformación Proteica en Lámina beta , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
PLoS One ; 12(6): e0179881, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28651013

RESUMEN

The cellular prion protein (PrPC) has been extensively studied because of its pivotal role in prion diseases; however, its functions remain incompletely understood. A unique line of goats has been identified that carries a nonsense mutation that abolishes synthesis of PrPC. In these animals, the PrP-encoding mRNA is rapidly degraded. Goats without PrPC are valuable in re-addressing loss-of-function phenotypes observed in Prnp knockout mice. As PrPC has been ascribed various roles in immune cells, we analyzed transcriptomic responses to loss of PrPC in peripheral blood mononuclear cells (PBMCs) from normal goat kids (n = 8, PRNP+/+) and goat kids without PrPC (n = 8, PRNPTer/Ter) by mRNA sequencing. PBMCs normally express moderate levels of PrPC. The vast majority of genes were similarly expressed in the two groups. However, a curated list of 86 differentially expressed genes delineated the two genotypes. About 70% of these were classified as interferon-responsive genes. In goats without PrPC, the majority of type I interferon-responsive genes were in a primed, modestly upregulated state, with fold changes ranging from 1.4 to 3.7. Among these were ISG15, DDX58 (RIG-1), MX1, MX2, OAS1, OAS2 and DRAM1, all of which have important roles in pathogen defense, cell proliferation, apoptosis, immunomodulation and DNA damage response. Our data suggest that PrPC contributes to the fine-tuning of resting state PBMCs expression level of type I interferon-responsive genes. The molecular mechanism by which this is achieved will be an important topic for further research into PrPC physiology.


Asunto(s)
Cabras/genética , Cabras/inmunología , Interferón Tipo I/genética , Proteínas PrPC/deficiencia , Animales , Línea Celular , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/genética , Humanos , Leucocitos/inmunología , Masculino , Ratones , Proteínas PrPC/genética , Proteínas PrPC/inmunología
6.
Biol Chem ; 397(11): 1115-1124, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27279060

RESUMEN

A number of unexpected pathophysiological connections linking different neurodegenerative diseases have emerged over the past decade. An example is provided by prion and Alzheimer's diseases. Despite being distinct pathologies, these disorders share several neurotoxic mechanisms, including accumulation of misfolded protein isoforms, stress of the protein synthesis machinery, and activation of a neurotoxic signaling mediated by the cellular prion protein. Here, in addition to reviewing these mechanisms, we will discuss the potential therapeutic interventions for prion and Alzheimer's diseases that are arising from the comprehension of their common neurodegenerative pathways.


Asunto(s)
Enfermedad de Alzheimer/terapia , Enfermedades por Prión/terapia , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Silenciador del Gen , Humanos , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Sci Rep ; 5: 17146, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26679898

RESUMEN

Although conversion of the cellular form of the prion protein (PrP(C)) into a misfolded isoform is the underlying cause of prion diseases, understanding PrP(C) physiological functions has remained challenging. PrP(C) depletion or overexpression alters the proliferation and differentiation properties of various types of stem and progenitor cells in vitro by unknown mechanisms. Such involvement remains uncertain in vivo in the absence of any drastic phenotype of mice lacking PrP(C). Here, we report PrP(C) enrichment at the base of the primary cilium in stem and progenitor cells from the central nervous system and cardiovascular system of developing mouse embryos. PrP(C) depletion in a neuroepithelial cell line dramatically altered key cilium-dependent processes, such as Sonic hedgehog signalling and α-tubulin post-translational modifications. These processes were also affected over a limited time window in PrP(C)-ablated embryos. Thus, our study reveals PrP(C) as a potential actor in the developmental regulation of microtubule dynamics and ciliary functions.


Asunto(s)
Cilios/metabolismo , Priones/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Células Cultivadas , Sistema Nervioso Central/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Proteínas Hedgehog/metabolismo , Ratones , Microscopía Confocal , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Priones/genética , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
8.
Cardiovasc Res ; 104(1): 93-102, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25139744

RESUMEN

AIMS: The cellular prion protein, PrP(C), whose aberrant isoforms are related to prion diseases of humans and animals, has a still obscure physiological function. Having observed an increased expression of PrP(C) in two in vivo paradigms of heart remodelling, we focused on isolated mouse hearts to ascertain the capacity of PrP(C) to antagonize oxidative damage induced by ischaemic and non-ischaemic protocols. METHODS AND RESULTS: Hearts isolated from mice expressing PrP(C) in variable amounts were subjected to different and complementary oxidative perfusion protocols. Accumulation of reactive oxygen species, oxidation of myofibrillar proteins, and cell death were evaluated. We found that overexpressed PrP(C) reduced oxidative stress and cell death caused by post-ischaemic reperfusion. Conversely, deletion of PrP(C) increased oxidative stress during both ischaemic preconditioning and perfusion (15 min) with H2O2. Supporting its relation with intracellular systems involved in oxidative stress, PrP(C) was found to influence the activity of catalase and, for the first time, the expression of p66(Shc), a protein implicated in oxidative stress-mediated cell death. CONCLUSIONS: Our data demonstrate that PrP(C) contributes to the cardiac mechanisms antagonizing oxidative insults.


Asunto(s)
Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Proteínas PrPC/metabolismo , Animales , Catalasa/metabolismo , Muerte Celular , Modelos Animales de Enfermedad , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Conejos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Factores de Tiempo
9.
Behav Brain Res ; 271: 325-32, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24975422

RESUMEN

Chronic consumption of drugs with addictive potential induces profound synaptic changes in the dopaminergic mesocorticolimbic pathway that underlie the long-term behavioral alterations seen in addicted subjects. Thus, exploring modulation systems of dopaminergic function may reveal novel targets to interfere with drug addiction. We recently showed that cellular prion protein (PrP(C)) affects the homeostasis of the dopaminergic system by interfering with dopamine synthesis, content, receptor density and signaling pathways in different brain areas. Here we report that the genetic deletion of PrP(C) modulates ethanol (EtOH)-induced behavioral alterations including the maintenance of drug seeking, voluntary consumption and the development of EtOH tolerance, all pivotal steps in drug addiction. Notably, these behavioral changes were accompanied by a significant depletion of dopamine levels in the prefrontal cortex and reduced dopamine D1 receptors in PrP(C) knockout mice. Furthermore, the pharmacological blockade of dopamine D1 receptors, but not D2 receptors, attenuated the abnormal EtOH consumption in PrP(C) knockout mice. Altogether, these findings provide new evidence that the PrP(C)/dopamine interaction plays a pivotal role in EtOH addictive properties in mice.


Asunto(s)
Adaptación Psicológica/efectos de los fármacos , Consumo de Bebidas Alcohólicas/psicología , Dopamina/deficiencia , Etanol/farmacología , Proteínas PrPC/deficiencia , Consumo de Bebidas Alcohólicas/genética , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
10.
Neurogastroenterol Motil ; 24(9): e412-24, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22762267

RESUMEN

BACKGROUND: Cellular prion protein (PrP(C) ) is expressed in the enteric nervous system (ENS), however, its physiological role has not been identified. Studies suggest that PrP(C) can function as a metal-binding protein, as absence of the protein has been linked to altered copper metabolism and atypical synaptic activity. Because copper is known to modulate smooth muscle relaxation, we tested the hypothesis that PrP(C) deficiency would alter intestinal contractility. METHODS: We examined electrically evoked ileal contractility in Prnp(-/-) or wild type littermate mice and the effects of copper or copper chelation. PrP(C) expression was studied in whole mount ileal preparations of mice and guinea pigs by immunohistochemistry. KEY RESULTS: Relative to wild type mice, ileal tissues of Prnp(-/-) mice exhibited reduced electrical field stimulation (EFS)-evoked contractility. Furthermore, EFS-induced relaxation, as a percentage of that induced by a nitric oxide donor, was enhanced. Addition of a copper donor to the organ bath increased, whereas the addition of a copper chelator inhibited, nitric oxide donor-induced ileal relaxation in Prnp(-/-) mice. PrP(C) was expressed on nerve fibers or terminals, and some cell bodies in the myenteric and submucosal plexuses of wild type mice. PrP(C) colocalized with a neuron-specific ectonucleotidase, nucleoside triphosphate diphosphohydrolase 3 (NTPDase3), but to only a limited extent with GFAP, a marker of enteric glia. Guinea pigs expressed PrP(C) in nerve fibers or terminals and enteric glia in the myenteric and submucosal plexuses. CONCLUSIONS & INFERENCES: Our findings suggest that PrP(C) , which is abundant in the ENS, has a role in the regulation of ileal contractility.


Asunto(s)
Cobre/fisiología , Íleon/fisiología , Contracción Muscular/fisiología , Relajación Muscular/fisiología , Músculo Liso/fisiología , Proteínas PrPC/fisiología , Animales , Quelantes , Cobre/metabolismo , Sistema Nervioso Entérico/fisiología , Cobayas , Íleon/inervación , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Músculo Liso/inervación , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas PrPC/deficiencia , Proteínas PrPC/metabolismo
11.
J Neuroinflammation ; 9: 25, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22281016

RESUMEN

BACKGROUND: The cellular prion protein (PrPc) is a host-encoded glycoprotein whose transconformation into PrP scrapie (PrPSc) initiates prion diseases. The role of PrPc in health is still obscure, but many candidate functions have been attributed to the protein, both in the immune and the nervous systems. Recent data show that experimental autoimmune encephalomyelitis (EAE) is worsened in mice lacking PrPc. Disease exacerbation has been attributed to T cells that would differentiate into more aggressive effectors when deprived of PrPc. However, alternative interpretations such as reduced resistance of neurons to autoimmune insult and exacerbated gliosis leading to neuronal deficits were not considered. METHOD: To better discriminate the contribution of immune cells versus neural cells, reciprocal bone marrow chimeras with differential expression of PrPc in the lymphoid or in the central nervous system (CNS) were generated. Mice were subsequently challenged with MOG35-55 peptide and clinical disease as well as histopathology were compared in both groups. Furthermore, to test directly the T cell hypothesis, we compared the encephalitogenicity of adoptively transferred PrPc-deficient versus PrPc-sufficient, anti-MOG T cells. RESULTS: First, EAE exacerbation in PrPc-deficient mice was confirmed. Irradiation exacerbated EAE in all the chimeras and controls, but disease was more severe in mice with a PrPc-deleted CNS and a normal immune system than in the reciprocal construction. Moreover, there was no indication that anti-MOG responses were different in PrPc-sufficient and PrPc-deficient mice. Paradoxically, PrPc-deficient anti-MOG 2D2 T cells were less pathogenic than PrPc-expressing 2D2 T cells. CONCLUSIONS: In view of the present data, it can be concluded that the origin of EAE exacerbation in PrPc-ablated mice resides in the absence of the prion protein in the CNS. Furthermore, the absence of PrPc on both neural and immune cells does not synergize for disease worsening. These conclusions highlight the critical role of PrPc in maintaining the integrity of the CNS in situations of stress, especially during a neuroinflammatory insult.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental , Neuronas/patología , Proteínas PrPC/deficiencia , Animales , Axones/patología , Linfocitos T CD4-Positivos/patología , Sistema Nervioso Central/efectos de la radiación , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Citometría de Flujo/métodos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicoproteínas/efectos adversos , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/efectos adversos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Factores de Tiempo , Irradiación Corporal Total
12.
Neuroscience ; 200: 130-41, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22062133

RESUMEN

Microglial activation is a key event in the progression and infiltration of tumors. We have previously demonstrated that the co-chaperone stress inducible protein 1 (STI1), a cellular prion protein (PrP(C)) ligand, promotes glioblastoma (GBM) proliferation. In the present study, we examined the influence of microglial STI1 in the growth and invasion of the human glioblastoma cell line GBM95. We demonstrated that soluble factors secreted by microglia into the culture medium (microglia conditioned medium; MG CM) caused a two-fold increase in the proliferation of GBM95 cells. This effect was reversed when STI1 was removed from the MG CM. In this context, we have shown that microglial cells synthesize and secrete STI1. Interestingly, no difference was observed in proliferation rates when GBM cells were maintained in MG CM or MG CM containing an anti-PrP(C) neutralizing antibody. Moreover, rec STI1 and rec STI1(Δ230-245), which lack the PrP(C) binding site, both promoted similar levels of GBM95 proliferation. In the migration assays, MG CM favored the migration of GBM95 cells, but migration failed when STI1 was removed from the MG CM. We detected metalloproteinase 9 (MMP-9) activity in the MG CM, and when cultured microglia were treated with an anti-STI1 antibody, MMP-9 activity decreased. Our results suggest that STI1 is secreted by microglia and favors tumor growth and invasion through the participation of MMP-9 in a PrP(C)-independent manner.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Glioblastoma/patología , Proteínas de Choque Térmico/farmacología , Microglía/química , Proteínas PrPC/metabolismo , Animales , Animales Recién Nacidos , Movimiento Celular/fisiología , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/química , Ratones , Ratones Noqueados , Neuronas/química , Proteínas PrPC/deficiencia , Timidina/metabolismo , Factores de Tiempo , Tritio/metabolismo
13.
J Proteome Res ; 11(2): 523-36, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22023170

RESUMEN

Approximately 25% of eukaryotic proteins possessing homology to at least two transmembrane domains are predicted to be embedded in biological membranes. Nevertheless, this group of proteins is not usually well represented in proteome-wide experiments due to their refractory nature. Here we present a quantitative mass spectrometry-based comparison of membrane protein expression in cerebellar granule neurons grown in primary culture that were isolated from wild-type mice and mice lacking the cellular prion protein. This protein is a cell-surface glycoprotein that is mainly expressed in the central nervous system and is involved in several neurodegenerative disorders, though its physiological role is unclear. We used a low specificity enzyme α-chymotrypsin to digest membrane proteins preparations that had been separated by SDS-PAGE. The resulting peptides were labeled with tandem mass tags and analyzed by MS. The differentially expressed proteins identified using this approach were further analyzed by multiple reaction monitoring to confirm the expression level changes.


Asunto(s)
Cerebelo/química , Proteínas de la Membrana/análisis , Neuronas/química , Proteínas PrPC/deficiencia , Proteoma/análisis , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Electroforesis en Gel de Poliacrilamida , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/metabolismo , Fragmentos de Péptidos/análisis , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteómica , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem
14.
J Neurochem ; 116(5): 881-90, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21214552

RESUMEN

The cellular prion protein (PrP(C)) is a cell-surface glycoprotein mainly expressed in the CNS. The structural conversion of PrP(C) generates the prion, the infectious agent causing transmissible spongiform encephalopathies, which are rare and fatal diseases affecting animals and humans. Despite decades of intensive research, the mechanism of prion-associated neurodegeneration and the physiologic role of PrP(C) are still obscure. Recent evidence, however, supports the hypothesis that PrP(C) may be involved in the control of Ca(2+) homeostasis. Given the universal significance of Ca(2+) as an intracellular messenger for both the life and death of cells, this possibility may help explain the complex, often controversial, dataset accumulated on PrP(C) physiology, and the events leading to prion-associated neuronal demise. In this study, we have compared local Ca(2+) movements in cerebellar granule neurons (CGN) derived from wild-type (WT), or PrP-knockout (KO), mice, by means of the Ca(2+)-sensitive photo-probe, aequorin, genetically targeted to specific intracellular domains and delivered to CGN by lentiviral vectors. The use of an aequorin that localizes to the cytosolic domains proximal to the plasma membrane has allowed us to demonstrate that there was a dramatic increase of store-operated Ca(2+) entry in PrP-KO CGN compared to WT neurons. Notably, this phenotype was rescued upon restoring PrP(C) expression. The Ca(2+)-phenotype of PrP-KO neurons can in part be explained by the lower expression of two major Ca(2+)-extruding proteins, namely the plasma membrane and the sarco-endoplasmic reticulum Ca(2+)-ATPases. The lower sarco-endoplasmic reticulum Ca(2+)-ATPase content may also contribute to explain why PrP-KO CGN accumulated less Ca(2+) in the endoplasmic reticulum than the WT counterpart.


Asunto(s)
Calcio/metabolismo , Cerebelo/citología , Neuronas/metabolismo , Proteínas PrPC/metabolismo , Priones/metabolismo , Aequorina/metabolismo , Animales , Anticuerpos/farmacología , Canales de Calcio/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ratones , Ratones Transgénicos , Neuronas/citología , Proteína ORAI2 , Proteínas PrPC/deficiencia , Proteínas PrPC/inmunología , Proteínas Priónicas , Priones/genética , Priones/inmunología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transfección/métodos
15.
Mol Cell Biol ; 30(20): 4864-76, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20679477

RESUMEN

It is now well established that the conversion of the cellular prion protein, PrP(C), into its anomalous conformer, PrP(Sc), is central to the onset of prion disease. However, both the mechanism of prion-related neurodegeneration and the physiologic role of PrP(C) are still unknown. The use of animal and cell models has suggested a number of putative functions for the protein, including cell signaling, adhesion, proliferation, and differentiation. Given that skeletal muscles express significant amounts of PrP(C) and have been related to PrP(C) pathophysiology, in the present study, we used skeletal muscles to analyze whether the protein plays a role in adult morphogenesis. We employed an in vivo paradigm that allowed us to compare the regeneration of acutely damaged hind-limb tibialis anterior muscles of mice expressing, or not expressing, PrP(C). Using morphometric and biochemical parameters, we provide compelling evidence that the absence of PrP(C) significantly slows the regeneration process compared to wild-type muscles by attenuating the stress-activated p38 pathway, and the consequent exit from the cell cycle, of myogenic precursor cells. Demonstrating the specificity of this finding, restoring PrP(C) expression completely rescued the muscle phenotype evidenced in the absence of PrP(C).


Asunto(s)
Músculo Esquelético/fisiología , Proteínas PrPC/fisiología , Regeneración/fisiología , Animales , Ciclo Celular , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/lesiones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Fenotipo , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Regeneración/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Nature ; 466(7308): E3-4; discussion E4-5, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20703260

RESUMEN

Increased levels of brain amyloid-beta, a secreted peptide cleavage product of amyloid precursor protein (APP), is believed to be critical in the aetiology of Alzheimer's disease. Increased amyloid-beta can cause synaptic depression, reduce the number of spine protrusions (that is, sites of synaptic contacts) and block long-term synaptic potentiation (LTP), a form of synaptic plasticity; however, the receptor through which amyloid-beta produces these synaptic perturbations has remained elusive. Laurén et al. suggested that binding between oligomeric amyloid-beta (a form of amyloid-beta thought to be most active) and the cellular prion protein (PrP(C)) is necessary for synaptic perturbations. Here we show that PrP(C) is not required for amyloid-beta-induced synaptic depression, reduction in spine density, or blockade of LTP; our results indicate that amyloid-beta-mediated synaptic defects do not require PrP(c).


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas PrPC/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Animales , Aprendizaje/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Reproducibilidad de los Resultados , Serotonina/metabolismo , Transmisión Sináptica
17.
Neuroscience ; 160(4): 731-43, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19285121

RESUMEN

The hypothesis of an early vulnerability of the serotonergic system to prion infection was investigated in a murine model of bovine spongiform encephalopathy (BSE). Behavioral tests targeted to 5-HT functions were performed in the course of infection to evaluate circadian activity, anxiety-like behavior, pain sensitivity and the 5-HT syndrome. The first behavioral change was a decrease in nocturnal activity detected at 30% of incubation time. Further behavioral alterations including nocturnal hyperactivity, reduced anxiety, hyperalgesia and exaggerated 5-HT syndrome were observed at 60%-70% of incubation time, before the onset of clinical signs. The same tests performed in 5-HT-depleted mice and in prion protein-deficient mice revealed behavioral abnormalities similar in many aspects to those of BSE-infected mice. Histological and biochemical analysis showed alterations of the serotonergic system in BSE-infected and prion protein-deficient mice. These results indicate that BSE infection affects the homeostasis of serotonergic neurons and suggest that the disruption of prion protein normal function contributes to the early pathological changes in our mouse model of BSE. A similar process may occur in the human variant Creutzfeldt-Jacob disease, as suggested by the early symptoms of alterations in mood, sleep and pain sensitivity.


Asunto(s)
Encéfalo/metabolismo , Encefalopatía Espongiforme Bovina/metabolismo , Trastornos Mentales/metabolismo , Proteínas PrPC/deficiencia , Proteínas PrPSc/toxicidad , Serotonina/metabolismo , Animales , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/fisiopatología , Encéfalo/fisiopatología , Tronco Encefálico/citología , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiopatología , Bovinos , Trastornos Cronobiológicos/genética , Trastornos Cronobiológicos/metabolismo , Trastornos Cronobiológicos/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalopatía Espongiforme Bovina/fisiopatología , Femenino , Homeostasis/fisiología , Trastornos Mentales/genética , Trastornos Mentales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Dolor/genética , Dolor/metabolismo , Dolor/fisiopatología , Proteínas PrPC/genética , Proteínas PrPSc/metabolismo , Núcleos del Rafe/citología , Núcleos del Rafe/metabolismo , Núcleos del Rafe/fisiopatología , Síndrome de la Serotonina/genética , Síndrome de la Serotonina/metabolismo , Síndrome de la Serotonina/fisiopatología , Factores de Tiempo
18.
Am J Pathol ; 173(4): 1029-41, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18815152

RESUMEN

Although the physiological roles of the cellular prion protein (PrP C) remain to be fully elucidated, PrP C has been proposed to represent a potential regulator of cellular immunity. To test this hypothesis, we evaluated the consequences of PrP C deficiency on the course of experimental autoimmune encephalomyelitis induced by immunization with myelin oligodendrocyte glycoprotein peptide. Consistent with augmented proliferative responses and increased cytokine gene expression by myelin oligodendrocyte glycoprotein-primed Prnp-/- T cells, PrP C-deficient mice demonstrated more aggressive disease onset and a lack of clinical improvement during the chronic phase of experimental autoimmune encephalomyelitis. Acutely, Prnp-/- spinal cord, cerebellum, and forebrain exhibited higher levels of leukocytic infiltrates and pro-inflammatory cytokine gene expression, as well as increased spinal cord myelin basic protein and axonal loss. During the chronic phase, a remarkable persistence of leukocytic infiltrates was present in the forebrain and cerebellum, accompanied by an increase in interferon-gamma and interleukin-17 transcripts. Attenuation of T cell-dependent neuroinflammation thus represents a potential novel function of PrP C.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Sistema Nervioso/patología , Proteínas PrPC/deficiencia , Animales , Conducta Animal , Linfocitos T CD4-Positivos/metabolismo , Cerebelo/patología , Reactividad Cruzada , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunización , Inflamación , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de la Mielina , Glicoproteína Asociada a Mielina , Glicoproteína Mielina-Oligodendrócito , Sistema Nervioso/metabolismo , Proteínas PrPC/metabolismo , Prosencéfalo/patología , Médula Espinal/patología , Regulación hacia Arriba
19.
Behav Brain Res ; 194(2): 129-37, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18590772

RESUMEN

The cellular prion protein (PrP(C)) is a neuronal anchored glycoprotein that has been associated with distinct functions in the CNS, such as cellular adhesion and differentiation, synaptic plasticity and cognition. Here we investigated the putative involvement of the PrP(C) in the innate fear-induced behavioural reactions in wild-type (WT), PrP(C) knockout (Prnp(0/0)) and the PrP(C) overexpressing Tg-20 mice evoked in a prey versus predator paradigm. The behavioural performance of these mouse strains in olfactory discrimination tasks was also investigated. When confronted with coral snakes, mice from both Prnp(0/0) and Tg-20 strains presented a significant decrease in frequency and duration of defensive attention and risk assessment, compared to WT mice. Tg-20 mice presented decreased frequency of escape responses, increased exploratory behaviour, and enhancement of interaction with the snake, suggesting a robust fearlessness caused by PrP(C) overexpression. Interestingly, there was also a discrete decrease in the attentional defensive response (decreased frequency of defensive alertness) in Prnp(0/0) mice in the presence of coral snakes. Moreover, Tg-20 mice presented an increased exploration of novel environment and odors. The present findings indicate that the PrP(C) overexpression causes hyperactivity, fearlessness, and increased preference for visual, tactile and olfactory stimuli-associated novelty, and that the PrP(c) deficiency might lead to attention deficits. These results suggest that PrP(c) exerts an important role in the modulation of innate fear and novelty-induced exploration.


Asunto(s)
Conducta Agonística/fisiología , Atención/fisiología , Miedo , Instinto , Proteínas PrPC/metabolismo , Análisis de Varianza , Animales , Conducta Animal , Elapidae , Reacción de Fuga/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas PrPC/deficiencia
20.
J Comp Neurol ; 508(3): 487-99, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18338400

RESUMEN

The normal physiological function of the prion protein PrP(C) remains elusive despite its widespread expression, particularly throughout the nervous system. A critical step toward identifying its function is to precisely localize its pattern of expression. Historically, the immunolocalization of PrP(C) has proved to be notoriously difficult and nonconsensual. We have thus undertaken a detailed expression analysis by means of a combination of in situ hybridization, knockout mice, and immunohistochemistry, using recently generated highly specific antibodies. We have attempted to accurately localize PrP(C) expression in a tissue that is highly structured and of crucial behavioral importance to mice, the olfactory system. We found that PrP(C) was expressed in both peripheral and central neurons of the olfactory system and that its distribution was axonal-specific in both olfactory sensory neurons of the olfactory epithelium and mitral cells of the olfactory bulb. Our detailed expression analysis and the axonal localization we observed may provide important hints toward potential functions of PrP(C).


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Expresión Génica/fisiología , Vías Olfatorias/metabolismo , Proteínas PrPC/metabolismo , Animales , Animales Recién Nacidos , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Noqueados , Neuronas Aferentes/metabolismo , Vías Olfatorias/citología , Proteínas PrPC/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...