Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Int J Mol Sci ; 22(11)2021 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-34071094

RESUMEN

Three main approaches are used to combat severe viral respiratory infections. The first is preemptive vaccination that blocks infection. Weakened or dead viral particles, as well as genetic constructs carrying viral proteins or information about them, are used as an antigen. However, the viral genome is very evolutionary labile and changes continuously. Second, chemical agents are used during infection and inhibit the function of a number of viral proteins. However, these drugs lose their effectiveness because the virus can rapidly acquire resistance to them. The third is the search for points in the host metabolism the effect on which would suppress the replication of the virus but would not have a significant effect on the metabolism of the host. Here, we consider the possibility of using the copper metabolic system as a target to reduce the severity of influenza infection. This is facilitated by the fact that, in mammals, copper status can be rapidly reduced by silver nanoparticles and restored after their cancellation.


Asunto(s)
Cobre/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Ceruloplasmina/fisiología , Proteínas Transportadoras de Cobre/metabolismo , ATPasas Transportadoras de Cobre/fisiología , Farmacorresistencia Viral , Interacciones Huésped-Patógeno , Humanos , Vacunas contra la Influenza , Gripe Humana/tratamiento farmacológico , Gripe Humana/prevención & control , Gripe Humana/virología , Mamíferos/metabolismo , Nanopartículas del Metal/uso terapéutico , Chaperonas Moleculares/metabolismo , Proteínas PrPC/fisiología , ARN Viral/fisiología , Plata/uso terapéutico , Superóxido Dismutasa-1/fisiología , Proteínas Virales/fisiología , Replicación Viral
2.
Acta Neuropathol ; 142(1): 5-16, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33899132

RESUMEN

Prions are novel pathogens that are composed entirely of PrPSc, the self-templating conformation of the host prion protein, PrPC. Prion strains are operationally defined as a heritable phenotype of disease that are encoded by strain-specific conformations of PrPSc. The factors that influence the relative distribution of strains in a population are only beginning to be understood. For prions with an infectious etiology, environmental factors, such as strain-specific binding to surfaces and resistance to weathering, can influence which strains are available for transmission to a naïve host. Strain-specific differences in efficiency of infection by natural routes of infection can also select for prion strains. The host amino acid sequence of PrPC has the greatest effect on dictating the repertoire of prion strains. The relative abundance of PrPC, post-translational modifications of PrPC and cellular co-factors involved in prion conversion can also provide conditions that favor the prevalence of a subset of prion strains. Additionally, prion strains can interfere with each other, influencing the emergence of a dominant strain. Overall, both environmental and host factors may influence the repertoire and distribution of strains within a population.


Asunto(s)
Evolución Biológica , Ambiente , Proteínas PrPC/genética , Proteínas PrPC/fisiología , Enfermedades por Prión/parasitología , Priones/genética , Priones/fisiología , Animales , Humanos , Proteínas PrPSc , Priones/clasificación
3.
Biomolecules ; 11(2)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33671884

RESUMEN

The cellular prion protein (PrPC) is a cell surface glycoprotein expressed in many cell types that plays an important role in normal cellular processes. However, an increase in PrPC expression has been associated with a variety of human cancers, where it may be involved in resistance to the proliferation and metastasis of cancer cells. PrP-deficient (Prnp0/0) and PrP-overexpressing (Tga20) mice were studied to evaluate the role of PrPC in the invasion and metastasis of cancer. Tga20 mice, with increased PrPC, died more quickly from lung cancer than did the Prnp0/0 mice, and this effect was associated with increased transforming growth factor-beta (TGF-ß) and programmed death ligand-1 (PD-L1), which are important for the development and function of regulatory T (Treg) cells. The number of FoxP3+CD25+ Treg cells was increased in Tga20 mice compared to Prnp0/0 mice, but there was no significant difference in either natural killer or cytotoxic T cell numbers. In addition, mice infected with the ME7 scrapie strain had decreased numbers of Treg cells and decreased expression of TGF-ß and PD-L1. These results suggest that PrPC plays an important role in invasion and metastasis of cancer cells by inducing Treg cells through upregulation of TGF-ß and PD-L1 expression.


Asunto(s)
Neoplasias Pulmonares/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas PrPC/fisiología , Linfocitos T Reguladores/inmunología , Animales , Antígeno B7-H1/metabolismo , Humanos , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Crecimiento Transformador beta/metabolismo
4.
Curr Issues Mol Biol ; 36: 67-88, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31559969

RESUMEN

Lowering cellular prion protein (PrPC) levels in the brain is predicted to be a powerful therapeutic strategy for the prion disease. PrPC may act as an antiapoptotic agent by blocking some of the internal environmental factors that initiate apoptosis. Prion protein (PrP)-knockout methods provide powerful indications on the neuroprotective function of PrPC. Using PrPC-knockout cell lines, the inhibition of apoptosis through stress inducible protein1 (STI1) is mediated by PrPC-dependent superoxide dismutase (SOD) activation. Besides, PrP-knockout exhibited wide spread alterations of oscillatory activity in the olfactory bulb as well as altered paired-pulse plasticity at the dendrodendric synapse. Both the behavioural and electro-physiological phenotypes could be rescued by neuronal PrPC expression. Neuprotein Shadoo (Sho), similarly to PrPC, can prevent neuronal cell death induced by the expression of PrP△HD mutants, an artificial PrP mutant devoid of internal hydrophobic domain. Sho can efficiently protect cells against exito-toxin-induced cell death by glutamates. Sho and PrP seem to be dependent on similar domains, in particular N-terminal (N), and their internal hydrophobic domain. Sho△N and Sho△HD displayed a reduced stress-protective activity but are complex glycosylated and attached to the outer leaflet of the plasma membrane via glycosylphosphatidylinositol (GPI) anchor indicating that impaired activity is not due to incorrect cellular trafficking. In Sho, over-expressed mice showed large amyloid plaques not seen in wild-type mice. However, Shadoo is not a major modulator of abnormal prion protein (PrPSc) accumulation. Sho and PrP share a stress-protective activity. The ability to adopt a toxic conformation of PrPSc seems to be specific for PrP.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas PrPC/metabolismo , Animales , Apoptosis/genética , Encéfalo/patología , Proteínas Ligadas a GPI , Mutación con Ganancia de Función , Mutación con Pérdida de Función , Ratones , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/fisiología , Proteínas PrPSc/metabolismo , Dominios Proteicos , Transducción de Señal/genética , Estrés Fisiológico/genética
5.
PLoS Pathog ; 15(3): e1007662, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30908557

RESUMEN

The protein-only hypothesis predicts that infectious mammalian prions are composed solely of PrPSc, a misfolded conformer of the normal prion protein, PrPC. However, protein-only PrPSc preparations lack significant levels of prion infectivity, leading to the alternative hypothesis that cofactor molecules are required to form infectious prions. Here, we show that prions with parental strain properties and full specific infectivity can be restored from protein-only PrPSc in vitro. The restoration reaction is rapid, potent, and requires bank vole PrPC substrate, post-translational modifications, and cofactor molecules. To our knowledge, this represents the first report in which the essential properties of an infectious mammalian prion have been restored from pure PrP without adaptation. These findings provide evidence for a unified hypothesis of prion infectivity in which the global structure of protein-only PrPSc accurately stores latent infectious and strain information, but cofactor molecules control a reversible switch that unmasks biological infectivity.


Asunto(s)
Proteínas PrPSc/metabolismo , Proteínas PrPSc/patogenicidad , Priones/metabolismo , Animales , Arvicolinae , Enfermedades Transmisibles , Mamíferos , Proteínas PrPC/metabolismo , Proteínas PrPC/fisiología , Proteínas PrPSc/fisiología , Proteínas Priónicas/metabolismo , Proteínas Priónicas/fisiología , Priones/patogenicidad , Priones/fisiología , Procesamiento Proteico-Postraduccional
6.
PLoS Pathog ; 15(1): e1007520, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30608982

RESUMEN

The cellular prion protein (PrPC) is a cell surface glycoprotein attached to the membrane by a glycosylphosphatidylinositol (GPI)-anchor and plays a critical role in transmissible, neurodegenerative and fatal prion diseases. Alterations in membrane attachment influence PrPC-associated signaling, and the development of prion disease, yet our knowledge of the role of the GPI-anchor in localization, processing, and function of PrPC in vivo is limited We exchanged the PrPC GPI-anchor signal sequence of for that of Thy-1 (PrPCGPIThy-1) in cells and mice. We show that this modifies the GPI-anchor composition, which then lacks sialic acid, and that PrPCGPIThy-1 is preferentially localized in axons and is less prone to proteolytic shedding when compared to PrPC. Interestingly, after prion infection, mice expressing PrPCGPIThy-1 show a significant delay to terminal disease, a decrease of microglia/astrocyte activation, and altered MAPK signaling when compared to wild-type mice. Our results are the first to demonstrate in vivo, that the GPI-anchor signal sequence plays a fundamental role in the GPI-anchor composition, dictating the subcellular localization of a given protein and, in the case of PrPC, influencing the development of prion disease.


Asunto(s)
Glicosilfosfatidilinositoles/metabolismo , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Animales , Modelos Animales de Enfermedad , Glicosilfosfatidilinositoles/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ácido N-Acetilneuramínico/metabolismo , Proteínas PrPC/fisiología , Enfermedades por Prión/genética , Proteínas Priónicas/metabolismo , Priones/genética , Priones/metabolismo , Señales de Clasificación de Proteína/fisiología , Transporte de Proteínas/fisiología , Proteolisis , Transducción de Señal
7.
Mol Cell ; 72(3): 426-443.e12, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401430

RESUMEN

Protein phase separation by low-complexity, intrinsically disordered domains generates membraneless organelles and links to neurodegeneration. Cellular prion protein (PrPC) contains such domains, causes spongiform degeneration, and is a receptor for Alzheimer's amyloid-ß oligomers (Aßo). Here, we show that PrPC separates as a liquid phase, in which α-helical Thr become unfolded. At the cell surface, PrPC Lys residues interact with Aßo to create a hydrogel containing immobile Aßo and relatively mobile PrPC. The Aßo/PrP hydrogel has a well-defined stoichiometry and dissociates with excess Aßo. NMR studies of hydrogel PrPC reveal a distinct α-helical conformation for natively unfolded amino-terminal Gly and Ala residues. Aßo/PrP hydrogel traps signal-transducing mGluR5 on the plasma membrane. Recombinant PrPC extracts endogenous Aßo from human Alzheimer's soluble brain lysates into hydrogel, and a PrPC antagonist releases Aßo from endogenous brain hydrogel. Thus, coupled phase and conformational transitions of PrPC are driven by Aß species from Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Proteínas PrPC/química , Proteínas PrPC/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo , Células COS , Línea Celular , Membrana Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Hidrogeles , Imagen por Resonancia Magnética/métodos , Conformación Molecular , Neuronas , Priones/química , Priones/fisiología , Unión Proteica , Receptor del Glutamato Metabotropico 5 , Transducción de Señal
8.
Exp Eye Res ; 175: 1-13, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29859760

RESUMEN

Iron is an essential biometal in the aqueous humor, the principal source of nutrients for the avascular cornea and the lens. Here, we explored whether the ciliary body (CB), the source of aqueous humor, transports iron, and if the prion protein (PrPC) facilitates this process as in the outer retina. Using a combination of human, bovine, and mouse eyes as models, we report the expression of iron export proteins ferroportin and ceruloplasmin, and major iron uptake and storage proteins transferrin, transferrin receptor, and ferritin in the ciliary epithelium, indicating active exchange of iron at this site. Ferroportin and transferrin receptor are also expressed in the corneal endothelium. However, the relative expression of iron export and uptake proteins suggests export from the ciliary epithelium and import by corneal endothelium. In addition, abundant expression of PrPC, a ferrireductase that facilitates iron transport, is noted in pigmented and non-pigmented epithelium of the CB, posterior pigmented epithelium of the iris, corneal endothelium and epithelium, and lens epithelium. Notably, majority of PrPC in the ciliary epithelium is cleaved at the ß-site as in retinal pigment epithelial cells, suggesting a role in iron transport. Most of the PrPC in the cornea, however, is full-length, and susceptible to aggregation by intracerebrally inoculated PrP-scrapie, an infectious conformation of PrPC responsible for human and animal prion disorders. Soluble PrPC is present in the aqueous and vitreous humor, a provocative observation with significant implications. Together, these observations suggest independent cycling of iron in the anterior segment, and a prominent role of PrPC in this process. Aggregation of PrPC in the cornea of PrP-scrapie-infected animals raises the alarming possibility of transmission of animal prions through corneal abrasions.


Asunto(s)
Segmento Anterior del Ojo/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas PrPC/fisiología , Enfermedad Debilitante Crónica/metabolismo , Enfermedad Debilitante Crónica/transmisión , Animales , Transporte Biológico , Western Blotting , Proteínas de Transporte de Catión/metabolismo , Bovinos , Ceruloplasmina , Cuerpo Ciliar/metabolismo , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/metabolismo , Femenino , Ferritinas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Homeostasis/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Transferrina/metabolismo , Transferrina/metabolismo
9.
PLoS Pathog ; 14(5): e1007049, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29723291

RESUMEN

The cellular prion protein, designated PrPC, is a membrane glycoprotein expressed abundantly in brains and to a lesser extent in other tissues. Conformational conversion of PrPC into the amyloidogenic isoform is a key pathogenic event in prion diseases. However, the physiological functions of PrPC remain largely unknown, particularly in non-neuronal tissues. Here, we show that PrPC is expressed in lung epithelial cells, including alveolar type 1 and 2 cells and bronchiolar Clara cells. Compared with wild-type (WT) mice, PrPC-null mice (Prnp0/0) were highly susceptible to influenza A viruses (IAVs), with higher mortality. Infected Prnp0/0 lungs were severely injured, with higher inflammation and higher apoptosis of epithelial cells, and contained higher reactive oxygen species (ROS) than control WT lungs. Treatment with a ROS scavenger or an inhibitor of xanthine oxidase (XO), a major ROS-generating enzyme in IAV-infected lungs, rescued Prnp0/0 mice from the lethal infection with IAV. Moreover, Prnp0/0 mice transgenic for PrP with a deletion of the Cu-binding octapeptide repeat (OR) region, Tg(PrPΔOR)/Prnp0/0 mice, were also highly susceptible to IAV infection. These results indicate that PrPC has a protective role against lethal infection with IAVs through the Cu-binding OR region by reducing ROS in infected lungs. Cu content and the activity of anti-oxidant enzyme Cu/Zn-dependent superoxide dismutase, SOD1, were lower in Prnp0/0 and Tg(PrPΔOR)/Prnp0/0 lungs than in WT lungs. It is thus conceivable that PrPC functions to maintain Cu content and regulate SOD1 through the OR region in lungs, thereby reducing ROS in IAV-infected lungs and eventually protecting them from lethal infection with IAVs. Our current results highlight the role of PrPC in protection against IAV infection, and suggest that PrPC might be a novel target molecule for anti-influenza therapeutics.


Asunto(s)
Proteínas PrPC/metabolismo , Proteínas Priónicas/metabolismo , Animales , Encéfalo/patología , Cobre/metabolismo , Susceptibilidad a Enfermedades/metabolismo , Virus de la Influenza A/metabolismo , Virus de la Influenza A/patogenicidad , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Proteínas PrPC/fisiología , Enfermedades por Prión/metabolismo , Proteínas Priónicas/farmacología , Priones/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
10.
J Virol ; 92(1)2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046443

RESUMEN

Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is a key pathogenic event in prion diseases, including Creutzfeldt-Jakob disease in humans and scrapie and bovine spongiform encephalopathy (BSE) in animals. We previously reported that the octapeptide repeat (OR) region could be dispensable for converting PrPC into PrPSc after infection with RML prions. We demonstrated that mice transgenically expressing mouse PrP with deletion of the OR region on the PrP knockout background, designated Tg(PrPΔOR)/Prnp0/0 mice, did not show reduced susceptibility to RML scrapie prions, with abundant accumulation of PrPScΔOR in their brains. We show here that Tg(PrPΔOR)/Prnp0/0 mice were highly resistant to BSE prions, developing the disease with markedly elongated incubation times after infection with BSE prions. The conversion of PrPΔOR into PrPScΔOR was markedly delayed in their brains. These results suggest that the OR region may have a crucial role in the conversion of PrPC into PrPSc after infection with BSE prions. However, Tg(PrPΔOR)/Prnp0/0 mice remained susceptible to RML and 22L scrapie prions, developing the disease without elongated incubation times after infection with RML and 22L prions. PrPScΔOR accumulated only slightly less in the brains of RML- or 22L-infected Tg(PrPΔOR)/Prnp0/0 mice than PrPSc in control wild-type mice. Taken together, these results indicate that the OR region of PrPC could play a differential role in the pathogenesis of BSE prions and RML or 22L scrapie prions.IMPORTANCE Structure-function relationship studies of PrPC conformational conversion into PrPSc are worthwhile to understand the mechanism of the conversion of PrPC into PrPSc We show here that, by inoculating Tg(PrPΔOR)/Prnp0/0 mice with the three different strains of RML, 22L, and BSE prions, the OR region could play a differential role in the conversion of PrPC into PrPSc after infection with RML or 22L scrapie prions and BSE prions. PrPΔOR was efficiently converted into PrPScΔOR after infection with RML and 22L prions. However, the conversion of PrPΔOR into PrPScΔOR was markedly delayed after infection with BSE prions. Further investigation into the role of the OR region in the conversion of PrPC into PrPSc after infection with BSE prions might be helpful for understanding the pathogenesis of BSE prions.


Asunto(s)
Susceptibilidad a Enfermedades , Encefalopatía Espongiforme Bovina/fisiopatología , Proteínas PrPC/química , Proteínas PrPC/fisiología , Enfermedades por Prión/fisiopatología , Priones/patogenicidad , Animales , Encéfalo/patología , Bovinos , Encefalopatía Espongiforme Bovina/prevención & control , Humanos , Ratones , Ratones Transgénicos , Oligopéptidos/química , Oligopéptidos/genética , Proteínas PrPC/genética , Enfermedades por Prión/prevención & control , Priones/química , Priones/genética , Eliminación de Secuencia
11.
Proc Natl Acad Sci U S A ; 114(5): 1141-1146, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096357

RESUMEN

Adaptation of prions to new species is thought to reflect the capacity of the host-encoded cellular form of the prion protein (PrPC) to selectively propagate optimized prion conformations from larger ensembles generated in the species of origin. Here we describe an alternate replicative process, termed nonadaptive prion amplification (NAPA), in which dominant conformers bypass this requirement during particular interspecies transmissions. To model susceptibility of horses to prions, we produced transgenic (Tg) mice expressing cognate PrPC Although disease transmission to only a subset of infected TgEq indicated a significant barrier to EqPrPC conversion, the resulting horse prions unexpectedly failed to cause disease upon further passage to TgEq. TgD expressing deer PrPC was similarly refractory to deer prions from diseased TgD infected with mink prions. In both cases, the resulting prions transmitted to mice expressing PrPC from the species of prion origin, demonstrating that transmission barrier eradication of the originating prions was ephemeral and adaptation superficial in TgEq and TgD. Horse prions produced in vitro by protein misfolding cyclic amplification of mouse prions using horse PrPC also failed to infect TgEq but retained tropism for wild-type mice. Concordant patterns of neuropathology and prion deposition in susceptible mice infected with NAPA prions and the corresponding prion of origin confirmed preservation of strain properties. The comparable responses of both prion types to guanidine hydrochloride denaturation indicated this occurs because NAPA precludes selection of novel prion conformations. Our findings provide insights into mechanisms regulating interspecies prion transmission and a framework to reconcile puzzling epidemiological features of certain prion disorders.


Asunto(s)
Especificidad del Huésped/fisiología , Proteínas PrPC/fisiología , Enfermedades por Prión/transmisión , Enfermedades por Prión/veterinaria , Priones/fisiología , Animales , Ciervos , Guanidina/farmacología , Caballos , Ratones , Ratones Endogámicos C57BL , Proteínas PrPC/química , Proteínas PrPC/genética , Priones/química , Conformación Proteica , Desnaturalización Proteica , Conejos , Ovinos , Especificidad de la Especie , Relación Estructura-Actividad
12.
J Virol ; 90(23): 10660-10669, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27654300

RESUMEN

Bank vole is a rodent species that shows differential susceptibility to the experimental transmission of different prion strains. In this work, the transmission features of a panel of diverse prions with distinct origins were assayed both in bank vole expressing methionine at codon 109 (Bv109M) and in transgenic mice expressing physiological levels of bank vole PrPC (the BvPrP-Tg407 mouse line). This work is the first systematic comparison of the transmission features of a collection of prion isolates, representing a panel of diverse prion strains, in a transgenic-mouse model and in its natural counterpart. The results showed very similar transmission properties in both the natural species and the transgenic-mouse model, demonstrating the key role of the PrP amino acid sequence in prion transmission susceptibility. However, differences in the PrPSc types propagated by Bv109M and BvPrP-Tg407 suggest that host factors other than PrPC modulate prion strain features. IMPORTANCE: The differential susceptibility of bank voles to prion strains can be modeled in transgenic mice, suggesting that this selective susceptibility is controlled by the vole PrP sequence alone rather than by other species-specific factors. Differences in the phenotypes observed after prion transmissions in bank voles and in the transgenic mice suggest that host factors other than the PrPC sequence may affect the selection of the substrain replicating in the animal model.


Asunto(s)
Arvicolinae/genética , Arvicolinae/fisiología , Proteínas PrPC/patogenicidad , Enfermedades por Prión/etiología , Priones/patogenicidad , Animales , Encéfalo/fisiopatología , Bovinos , Síndrome de Creutzfeldt-Jakob/etiología , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/transmisión , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Transgénicos , Proteínas PrPC/genética , Proteínas PrPC/fisiología , Enfermedades por Prión/genética , Enfermedades por Prión/transmisión , Priones/genética , Priones/fisiología , Ovinos , Especificidad de la Especie
14.
Neurochem Res ; 41(7): 1691-9, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26975317

RESUMEN

Cellular prion protein (PrP(C)) is a glycoprotein of the plasma membrane that plays pleiotropic functions by interacting with multiple signaling complexes at the cell surface. Recently, a number of studies have reported the involvement of PrP(C) in dopamine metabolism and signaling, including its interactions with tyrosine hydroxylase (TH) and dopamine receptors. However, the outcomes reported by independent studies are still debatable. Therefore in this study, we investigated the effects of PrP(C) on the TH expression during the differentiation of N2a cells with dibutyryl-cAMP, a well-known cAMP analog that activates TH transcription. Upon differentiation, TH was induced with concomitant reduction of PrP(C) at protein level, but not at mRNA level. shRNA-mediated PrP(C) reduction increased the basal level of TH at both mRNA and protein levels without dibutyryl-cAMP treatment. This phenotype was reversed by re-expression of PrP(C). PrP(C) knockdown also potentiated the effect of dibutyryl-cAMP on TH expression. Our findings suggest that PrP(C) has suppressive effects on TH expression. As a consequence, altered PrP(C) functions may affect the regulation of dopamine metabolism and related neurological disorders.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteínas PrPC/fisiología , Tirosina 3-Monooxigenasa/biosíntesis , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Dopamina/metabolismo , Ratones , Tirosina 3-Monooxigenasa/genética
15.
Cell Mol Life Sci ; 73(3): 667-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26298290

RESUMEN

The cellular prion protein (PrP(C)) is a ubiquitously expressed protein of currently unresolved but potentially diverse function. Of putative relevance to normal biological activity, PrP(C) is recognized to undergo both α- and ß-endoproteolysis, producing the cleavage fragment pairs N1/C1 and N2/C2, respectively. Experimental evidence suggests the likelihood that these processing events serve differing cellular needs. Through the engineering of a C-terminal c-myc tag onto murine PrP(C), as well as the selective use of a far-C-terminal anti-PrP antibody, we have identified a new PrP(C) fragment, nominally 'C3', and elaborating existing nomenclature, 'γ-cleavage' as the responsible proteolysis. Our studies indicate that this novel γ-cleavage event can occur during transit through the secretory pathway after exiting the endoplasmic reticulum, and after PrP(C) has reached the cell surface, by a matrix metalloprotease. We found that C3 is GPI-anchored like other C-terminal and full length PrP(C) species, though it does not localize primarily at the cell surface, and is preferentially cleaved from an unglycosylated substrate. Importantly, we observed that C3 exists in diverse cell types as well as mouse and human brain tissue, and of possible pathogenic significance, γ-cleavage may increase in human prion diseases. Given the likely relevance of PrP(C) processing to both its normal function, and susceptibility to prion disease, the potential importance of this previously underappreciated and overlooked cleavage event warrants further consideration.


Asunto(s)
Fragmentos de Péptidos/fisiología , Proteínas PrPC/metabolismo , Animales , Línea Celular , Humanos , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/fisiología , Ratones , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteínas PrPC/química , Proteínas PrPC/fisiología , Enfermedades por Prión/metabolismo , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Proteolisis
16.
J Neurochem ; 137(1): 88-100, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26710111

RESUMEN

Alzheimer's disease is a common neurodegenerative, progressive, and fatal disorder. Generation and deposition of amyloid beta (Aß) peptides associate with its pathogenesis and small soluble Aß oligomers show the most pronounced neurotoxic effects and correlate with disease initiation and progression. Recent findings showed that Aß oligomers bind to the cellular prion protein (PrP(C) ) eliciting neurotoxic effects. The role of exosomes, small extracellular vesicles of endosomal origin, in Alzheimer's disease is only poorly understood. Besides serving as disease biomarkers they may promote Aß plaque formation, decrease Aß-mediated synaptotoxicity, and enhance Aß clearance. Here, we explore how exosomal PrP(C) connects to protective functions attributed to exosomes in Alzheimer's disease. To achieve this, we generated a mouse neuroblastoma PrP(C) knockout cell line using transcription activator-like effector nucleases. Using these, as well as SH-SY5Y human neuroblastoma cells, we show that PrP(C) is highly enriched on exosomes and that exosomes bind amyloid beta via PrP(C) . Exosomes showed highest binding affinity for dimeric, pentameric, and oligomeric Aß species. Thioflavin T assays revealed that exosomal PrP(C) accelerates fibrillization of amyloid beta, thereby reducing neurotoxic effects imparted by oligomeric Aß. Our study provides further evidence for a protective role of exosomes in Aß-mediated neurodegeneration and highlights the importance of exosomal PrP(C) in molecular mechanisms of Alzheimer's disease. We show that the prion protein (PrP(C) ) on exosomes captures neurotoxic species of amyloid beta (Aß) promoting its fibrillization. Our study provides evidence for a protective role of exosomes in Alzheimer`s disease and suggests that, depending on its membrane topology, PrP(C) holds a dual function: when expressed at the neuronal surface it acts as receptor for Aß leading to neurotoxic signaling, whereas it detoxifies Aß when present on exosomes. This provides further support for key roles of PrP(C) in Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Exosomas/fisiología , Proteínas PrPC/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Ratones , Proteínas de Neoplasias/metabolismo , Neuroblastoma/patología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Solubilidad , Transfección
19.
J Biol Chem ; 290(33): 20488-98, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26152722

RESUMEN

We sought to examine interactions of the prion protein (PrP(C)) with monoaminergic systems due to: the role of PrP(C) in both Prion and Alzheimer diseases, which include clinical depression among their symptoms, the implication of monoamines in depression, and the hypothesis that PrP(C) serves as a scaffold for signaling systems. To that effect we compared both behavior and monoaminergic markers in wild type (WT) and PrP(C)-null (PrP(-/-)) mice. PrP(-/-) mice performed poorly when compared with WT in forced swimming, tail suspension, and novelty suppressed feeding tests, typical of depressive-like behavior, but not in the control open field nor rotarod motor tests; cyclic AMP responses to stimulation of D1 receptors by dopamine was selectively impaired in PrP(-/-) mice, and responses to serotonin, but not to norepinephrine, also differed between genotypes. Contents of dopamine, tyrosine hydroxylase, and the 5-HT5A serotonin receptor were increased in the cerebral cortex of PrP(-/-), as compared with WT mice. Microscopic colocalization, as well as binding in overlay assays were found of PrP(C) with both the 5HT5A and D1, but not D4 receptors. The data are consistent with the scaffolding of monoaminergic signaling modules by PrP(C), and may help understand the pathogenesis of clinical depression and neurodegenerative disorders.


Asunto(s)
Conducta Animal , Monoaminas Biogénicas/fisiología , Depresión/fisiopatología , Proteínas PrPC/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas PrPC/genética
20.
Biochem Biophys Res Commun ; 464(2): 512-8, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26159919

RESUMEN

Cellular prion protein (PrP(C)) is widely expressed in various cell types, including cells of the immune system. However, the specific roles of PrP(C) in the immune system have not been clearly elucidated. In the present study, we investigated the effects of a soluble form of recombinant PrP(C) protein on human natural killer (NK) cells. Recombinant soluble PrP(C) protein was generated by fusion of human PrP(C) with the Fc portion of human IgG1 (PrP(C)-Fc). PrP(C)-Fc binds to the surface of human NK cells, particularly to CD56(dim) NK cells. PrP(C)-Fc induced the production of cytokines and chemokines and the degranulation of granzyme B from NK cells. In addition, PrP(C)-Fc facilitated the IL-15-induced proliferation of NK cells. PrP(C)-Fc induced phosphorylation of ERK-1/2 and JNK in NK cells, and inhibitors of the ERK or the JNK pathways abrogated PrP(C)-Fc-induced cytokine production in NK cells. In conclusion, the soluble form of recombinant PrP(C)-Fc protein activates human NK cells via the ERK and JNK signaling pathways.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/fisiología , Proteínas PrPC/fisiología , Degranulación de la Célula/fisiología , Células Cultivadas , Granzimas/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...