Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Biol Chem ; 294(37): 13619-13628, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31320473

RESUMEN

Prion diseases are a group of incurable neurodegenerative disorders that affect humans and animals via infection with proteinaceous particles called prions. Prions are composed of PrPSc, a misfolded version of the cellular prion protein (PrPC). During disease progression, PrPSc replicates by interacting with PrPC and inducing its conversion to PrPSc As PrPSc accumulates, cellular stress mechanisms are activated to maintain cellular proteostasis, including increased protein chaperone levels. However, the exact roles of several of these chaperones remain unclear. Here, using various methodologies to monitor prion replication (i.e. protein misfolding cyclic amplification and cellular and animal infectivity bioassays), we studied the potential role of the molecular chaperone heat shock protein 70 (HSP70) in prion replication in vitro and in vivo Our results indicated that pharmacological induction of the heat shock response in cells chronically infected with prions significantly decreased PrPSc accumulation. We also found that HSP70 alters prion replication in vitro More importantly, prion infection of mice lacking the genes encoding stress-induced HSP70 exhibited accelerated prion disease progression compared with WT mice. In parallel with HSP70 being known to respond to endogenous and exogenous stressors such as heat, infection, toxicants, and ischemia, our results indicate that HSP70 may also play an important role in suppressing or delaying prion disease progression, opening opportunities for therapeutic intervention.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Enfermedades por Prión/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Progresión de la Enfermedad , Retículo Endoplásmico/fisiología , Estrés del Retículo Endoplásmico/fisiología , Femenino , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Priónicas/metabolismo , Priones/metabolismo , Pliegue de Proteína
2.
PLoS Pathog ; 14(1): e1006826, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29338055

RESUMEN

To explore pathogenesis in a young Gerstmann-Sträussler-Scheinker Disease (GSS) patient, the corresponding mutation, an eight-residue duplication in the hydrophobic region (HR), was inserted into the wild type mouse PrP gene. Transgenic (Tg) mouse lines expressing this mutation (Tg.HRdup) developed spontaneous neurologic syndromes and brain extracts hastened disease in low-expressor Tg.HRdup mice, suggesting de novo formation of prions. While Tg.HRdup mice exhibited spongiform change, PrP aggregates and the anticipated GSS hallmark of a proteinase K (PK)-resistant 8 kDa fragment deriving from the center of PrP, the LGGLGGYV insertion also imparted alterations in PrP's unstructured N-terminus, resulting in a 16 kDa species following thermolysin exposure. This species comprises a plausible precursor to the 8 kDa PK-resistant fragment and its detection in adolescent Tg.HRdup mice suggests that an early start to accumulation could account for early disease of the index case. A 16 kDa thermolysin-resistant signature was also found in GSS patients with P102L, A117V, H187R and F198S alleles and has coordinates similar to GSS stop codon mutations. Our data suggest a novel shared pathway of GSS pathogenesis that is fundamentally distinct from that producing structural alterations in the C-terminus of PrP, as observed in other prion diseases such as Creutzfeldt-Jakob Disease and scrapie.


Asunto(s)
Enfermedad de Gerstmann-Straussler-Scheinker/genética , Mutación , Proteínas PrPSc/química , Proteínas PrPSc/genética , Enfermedades por Prión/genética , Adulto , Alelos , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fragmentos de Péptidos/genética , Proteínas PrPSc/metabolismo , Dominios Proteicos/genética , Precursores de Proteínas/química , Precursores de Proteínas/genética
3.
Mol Psychiatry ; 23(12): 2363-2374, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171212

RESUMEN

The typical abnormalities observed in the brain of Alzheimer's disease (AD) patients include synaptic alterations, neuronal death, brain inflammation, and the accumulation of protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Despite the development of many animal and in vitro models for AD, there is a lack of an experimental approach that fully recapitulates essential aspects of the disease in human cells. Here, we report the generation of a new model to study AD, consisting of cerebral organoids (COs) produced from human-induced pluripotent stem cells (iPSCs). Under our experimental conditions, COs grow to form three-dimensional (3D) structures containing neural areas with cortical-like organization. Analysis of COs by histological and biochemical methods revealed that organoids produced from iPSCs derived from patients affected by familial AD or Down syndrome (DS) spontaneously develop over time pathological features of AD, including accumulation of structures highly reminiscent to amyloid plaques and neurofibrillary tangles. These pathological abnormalities were not observed in COs generated from various controls, including human iPSCs from healthy individuals, human iPSCs from patients affected by Creutzfeldt-Jakob disease, mouse embryonic stem cells (ESCs), or mouse iPSCs. These findings enable modeling genetic AD in a human cellular context in a 3D cortical-like tissue developed in vitro from patient-specific stem cells. This system provides a more relevant disease model compared to pre-existing methods and offers a new platform for discovery of novel targets and screening of drugs for therapeutic intervention.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Organoides/metabolismo , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/fisiología , Encéfalo/metabolismo , Técnicas de Cultivo de Célula/métodos , Corteza Cerebral , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Fosforilación , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/fisiología
4.
Cell Mol Life Sci ; 75(19): 3521-3538, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30030591

RESUMEN

Protein misfolding and aggregation into fibrillar deposits is a common feature of a large group of degenerative diseases affecting the central nervous system or peripheral organs, termed protein misfolding disorders (PMDs). Despite their established toxic nature, clinical trials aiming to reduce misfolded aggregates have been unsuccessful in treating or curing PMDs. An interesting possibility for disease intervention is the regular intake of natural food or herbal extracts, which contain active molecules that inhibit aggregation or induce the disassembly of misfolded aggregates. Among natural compounds, phenolic molecules are of particular interest, since most have dual activity as amyloid aggregation inhibitors and antioxidants. In this article, we review many phenolic natural compounds which have been reported in diverse model systems to have the potential to delay or prevent the development of various PMDs, including Alzheimer's and Parkinson's diseases, prion diseases, amyotrophic lateral sclerosis, systemic amyloidosis, and type 2 diabetes. The lower toxicity of natural compounds compared to synthetic chemical molecules suggest that they could serve as a good starting point to discover protein misfolding inhibitors that might be useful for the treatment of various incurable diseases.


Asunto(s)
Productos Biológicos/farmacología , Fenoles/farmacología , Agregación Patológica de Proteínas/prevención & control , Pliegue de Proteína/efectos de los fármacos , Deficiencias en la Proteostasis/prevención & control , Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Productos Biológicos/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Fenoles/uso terapéutico , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Deficiencias en la Proteostasis/tratamiento farmacológico
5.
J Virol ; 89(24): 12418-26, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423957

RESUMEN

UNLABELLED: In lethal prion neurodegenerative diseases, misfolded prion proteins (PrP(Sc)) replicate by redirecting the folding of the cellular prion glycoprotein (PrP(C)). Infections of different durations can have a subclinical phase with constant levels of infectious particles, but the mechanisms underlying this plateau and a subsequent exit to overt clinical disease are unknown. Using tandem biophysical techniques, we show that attenuated accumulation of infectious particles in presymptomatic disease is preceded by a progressive fall in PrP(C) level, which constricts replication rate and thereby causes the plateau effect. Furthermore, disease symptoms occurred at the threshold associated with increasing levels of small, relatively less protease-resistant oligomeric prion particles (oPrP(Sc)). Although a hypothetical lethal isoform of PrP cannot be excluded, our data argue that diminishing residual PrP(C) levels and continuously increasing levels of oPrP(Sc) are crucial determinants in the transition from presymptomatic to symptomatic prion disease. IMPORTANCE: Prions are infectious agents that cause lethal brain diseases; they arise from misfolding of a cell surface protein, PrP(C) to a form called PrP(Sc). Prion infections can have long latencies even though there is no protective immune response. Accumulation of infectious prion particles has been suggested to always reach the same plateau in the brain during latent periods, with clinical disease only occurring when hypothetical toxic forms (called PrP(L) or TPrP) begin to accumulate. We show here that infectivity plateaus arise because PrP(C) precursor levels become downregulated and that the duration of latent periods can be accounted for by the level of residual PrP(C), which transduces a toxic effect, along with the amount of oligomeric forms of PrP(Sc).


Asunto(s)
Encéfalo/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Animales , Encéfalo/patología , Ratones , Ratones Transgénicos , Proteínas PrPSc/genética , Enfermedades por Prión/genética , Enfermedades por Prión/patología
6.
PLoS Pathog ; 9(11): e1003755, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244171

RESUMEN

Prion diseases are driven by the strain-specific, template-dependent transconformation of the normal cellular prion protein (PrP(C)) into a disease specific isoform PrP(Sc). Cell culture models of prion infection generally use replicating cells resulting in lower levels of prion accumulation compared to animals. Using non-replicating cells allows the accumulation of higher levels of PrP(Sc) and, thus, greater amounts of infectivity. Here, we infect non-proliferating muscle fiber myotube cultures prepared from differentiated myoblasts. We demonstrate that prion-infected myotubes generate substantial amounts of PrP(Sc) and that the level of infectivity produced in these post-mitotic cells, 10(5.5) L.D.50/mg of total protein, approaches that observed in vivo. Exposure of the myotubes to different mouse-adapted agents demonstrates strain-specific replication of infectious agents. Mouse-derived myotubes could not be infected with hamster prions suggesting that the species barrier effect is intact. We suggest that non-proliferating myotubes will be a valuable model system for generating infectious prions and for screening compounds for anti-prion activity.


Asunto(s)
Proliferación Celular , Proteínas PrPSc/metabolismo , Animales , Línea Celular , Cricetinae , Ratones , Fibras Musculares Esqueléticas , Especificidad de la Especie
7.
PLoS Pathog ; 7(11): e1002391, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22114562

RESUMEN

During prion infections of the central nervous system (CNS) the cellular prion protein, PrP(C), is templated to a conformationally distinct form, PrP(Sc). Recent studies have demonstrated that the Sprn gene encodes a GPI-linked glycoprotein Shadoo (Sho), which localizes to a similar membrane environment as PrP(C) and is reduced in the brains of rodents with terminal prion disease. Here, analyses of prion-infected mice revealed that down-regulation of Sho protein was not related to Sprn mRNA abundance at any stage in prion infection. Down-regulation was robust upon propagation of a variety of prion strains in Prnp(a) and Prnp(b) mice, with the exception of the mouse-adapted BSE strain 301 V. In addition, Sho encoded by a TgSprn transgene was down-regulated to the same extent as endogenous Sho. Reduced Sho levels were not seen in a tauopathy, in chemically induced spongiform degeneration or in transgenic mice expressing the extracellular ADan amyloid peptide of familial Danish dementia. Insofar as prion-infected Prnp hemizygous mice exhibited accumulation of PrP(Sc) and down-regulation of Sho hundreds of days prior to onset of neurologic symptoms, Sho depletion can be excluded as an important trigger for clinical disease or as a simple consequence of neuronal damage. These studies instead define a disease-specific effect, and we hypothesize that membrane-associated Sho comprises a bystander substrate for processes degrading PrP(Sc). Thus, while protease-resistant PrP detected by in vitro digestion allows post mortem diagnosis, decreased levels of endogenous Sho may trace an early response to PrP(Sc) accumulation that operates in the CNS in vivo. This cellular response may offer new insights into the homeostatic mechanisms involved in detection and clearance of the misfolded proteins that drive prion disease pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas PrPSc/biosíntesis , Enfermedades por Prión/metabolismo , Animales , Regulación hacia Abajo , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas PrPC/metabolismo , ARN Mensajero/metabolismo
8.
Biochemistry ; 51(45): 9029-31, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23121093

RESUMEN

Shadoo (Sho) is a central nervous system glycoprotein with characteristics similar to those of the cellular prion protein PrP(C), each containing a highly conserved hydrophobic domain (HD) and an N-terminal repeat region. Whereas PrP(C) includes histidine-containing octarepeats, the Sho region N-terminal to the HD includes tandem positively charged "RGG boxes", predicted to bind RNA. Here, we demonstrate that Sho binds DNA and RNA in vitro via this arginine-rich region.


Asunto(s)
Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , ARN Bacteriano/metabolismo , Animales , Arginina/química , Proteínas Ligadas a GPI , Lisina/química , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Repeticiones de Trinucleótidos/fisiología
9.
Front Mol Neurosci ; 15: 990136, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36117913

RESUMEN

Many questions surround the underlying mechanism for the differential metabolic processing observed for the prion protein (PrP) in healthy and prion-infected mammals. Foremost, the physiological α-cleavage of PrP interrupts a region critical for both toxicity and conversion of cellular PrP (PrP C ) into its misfolded pathogenic isoform (PrP Sc ) by generating a glycosylphosphatidylinositol (GPI)-anchored C1 fragment. During prion diseases, alternative ß-cleavage of PrP becomes prominent, producing a GPI-anchored C2 fragment with this particular region intact. It remains unexplored whether physical up-regulation of α-cleavage can inhibit disease progression. Furthermore, several pieces of evidence indicate that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 play a much smaller role in the α-cleavage of PrP C than originally believed, thus presenting the need to identify the primary protease(s) responsible. For this purpose, we characterized the ability of plasmin to perform PrP α-cleavage. Then, we conducted functional assays using protein misfolding cyclic amplification (PMCA) and prion-infected cell lines to clarify the role of plasmin-mediated α-cleavage during prion propagation. Here, we demonstrated an inhibitory role of plasmin for PrP Sc formation through PrP α-cleavage that increased C1 fragments resulting in reduced prion conversion compared with non-treated PMCA and cell cultures. The reduction of prion infectious titer in the bioassay of plasmin-treated PMCA material also supported the inhibitory role of plasmin on PrP Sc replication. Our results suggest that plasmin-mediated endoproteolytic cleavage of PrP may be an important event to prevent prion propagation.

10.
FASEB J ; 24(12): 5102-12, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20732953

RESUMEN

To clarify the role of plasminogen as a cofactor for prion propagation, we conducted functional assays using a cell-free prion protein (PrP) conversion assay termed protein misfolding cyclic amplification (PMCA) and prion-infected cell lines. Here, we report that plasminogen stimulates propagation of the protease-resistant scrapie PrP (PrP(Sc)). Compared to control PMCA conducted without plasminogen, addition of plasminogen in PMCA using wild-type brain material significantly increased PrP conversion, with an EC(50) = ∼56 nM. PrP conversion in PMCA was substantially less efficient with plasminogen-deficient brain material than with wild-type material. The activity stimulating PrP conversion was specific for plasminogen and conserved in its kringle domains. Such activity was abrogated by modification of plasminogen structure and interference of PrP-plasminogen interaction. Kinetic analysis of PrP(Sc) generation demonstrated that the presence of plasminogen in PMCA enhanced the PrP(Sc) production rate to ∼0.97 U/µl/h and reduced turnover time to ∼1 h compared to those (∼0.4 U/µl/h and ∼2.5 h) obtained without supplementation. Furthermore, as observed in PMCA, plasminogen and kringles promoted PrP(Sc) propagation in ScN2a and Elk 21(+) cells. Our results demonstrate that plasminogen functions in stimulating conversion processes and represents the first cellular protein cofactor that enhances the hypothetical mechanism of prion propagation.


Asunto(s)
Plasminógeno/química , Plasminógeno/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo , Animales , Humanos , Técnicas In Vitro , Ratones , Unión Proteica/fisiología , Pliegue de Proteína
11.
Biochem Biophys Res Commun ; 388(2): 306-10, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19664595

RESUMEN

Protein misfolding cyclic amplification (PMCA) is a cell-free assay mimicking the prion replication process. However, constraints affecting PMCA have not been well-defined. Although cellular prion protein (PrP(C)) is required for prion replication, the influence of PrP(C) abundance on PMCA has not been assessed. Here, we show that PMCA was enhanced by using mouse brain material in which PrP(C) was overexpressed. Tg(MoPrP)4112 mice overexpressing PrP(C) supported more sensitive and efficient PMCA than wild type mice. As brain homogenate of Tg(MoPrP)4112 mice was diluted with PrP(C)-deficient brain material, PMCA became less robust. Our studies suggest that abundance of PrP(C) is a determinant that directs enhancement of PMCA. PMCA established here will contribute to optimizing conditions to enhance PrP(Sc) amplification by using concentrated PrP(C) source and expands the use of this methodology.


Asunto(s)
Proteínas PrPC/biosíntesis , Proteínas PrPC/química , Replicación de Secuencia Autosostenida/métodos , Animales , Encéfalo/metabolismo , Química Encefálica , Ratones , Proteínas PrPC/genética , Pliegue de Proteína
12.
Mol Cell Biochem ; 328(1-2): 177-82, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19322640

RESUMEN

Prion protein (PrP) interacts with some kringle domain-containing proteins. Kringle domains serve as binding domains in the interaction with PrP. The structural conservation among kringle domains leads to the hypothesis that any protein containing these domains can interact with PrP and be involved in prion pathogenesis. Because prion pathogenesis occurs in the brain, kringle domain-containing proteins should be available in the same tissue if they are relevant to prion pathogenesis. However, gene expression of these proteins in brains infected by prions has not been examined. Here, we showed that plasminogen (plg), urokinase type plasminogen activator (upa), tissue type plasminogen activator (tpa), prothrombin (prothr), and hepatocyte growth factor (hgf) genes were expressed in murine brains and neuroblastoma cells. The changes in upa, prothr, and hgf gene expression correlated with prion disease, but those in plg and tpa gene expression did not. Our data suggest association of gene expression of kringle domain-containing proteins in brains with prion disease.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Kringles , Neuroblastoma/metabolismo , Enfermedades por Prión/genética , Priones/metabolismo , Proteínas/genética , Animales , Sitios de Unión , Factor de Crecimiento de Hepatocito/genética , Ratones , Plasminógeno/genética , Unión Proteica/genética , Protrombina/genética , Activador de Tejido Plasminógeno/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética
13.
Sci Rep ; 9(1): 4847, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890734

RESUMEN

Advances in biotechnology have led to the development of a number of biological therapies for the treatment of diverse human diseases. Since these products may contain or are made using human or animal (e.g. cattle) derived materials, it is crucial to test their safety by ensuring the absence of infectious agents; specifically prions, which are highly resilient to elimination and produce fatal diseases in humans. Many cases of iatrogenic Creutzfeldt-Jakob disease have been caused by the use of biological materials (e.g. human growth hormone) contaminated with prions. For this reason, it is important to screen cells and biological materials for the presence of prions. Here we show the utility of the Protein Misfolding Cyclic Amplification (PMCA) technology as a screening tool for the presence of human (vCJD) and bovine (BSE) prions in a human cell therapy product candidate. First, we demonstrated the sensitivity of PMCA to detect a single cell infected with prions. For these experiments, we used RKM7 cells chronically infected with murine RML prions. Serial dilutions of an infected cell culture showed that PMCA enabled prion amplification from a sample comprised of only one cell. Next, we determined that PMCA performance was robust and uncompromised by the spiking of large quantities of uninfected cells into the reaction. Finally, to demonstrate the practical application of this technology, we analyzed a human cell line being developed for therapeutic use and found it to be PMCA-negative for vCJD and BSE prions. Our findings demonstrate that the PMCA technology has unparalleled sensitivity and specificity for the detection of prions, making it an ideal quality control procedure in the production of biological therapeutics.


Asunto(s)
Productos Biológicos/farmacología , Biotecnología/métodos , Síndrome de Creutzfeldt-Jakob/tratamiento farmacológico , Priones/efectos de los fármacos , Animales , Línea Celular , Humanos , Pliegue de Proteína/efectos de los fármacos , Conejos , Sensibilidad y Especificidad
14.
Mol Neurobiol ; 56(11): 7888-7904, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31129810

RESUMEN

The Shadoo protein (Sho) exhibits homology to the hydrophobic region of the cellular isoform of prion protein (PrPC). As prion-infected brains gradually accumulate infectivity-associated isoforms of prion protein (PrPSc), levels of mature endogenous Sho become reduced. To study the regulatory effect of the proteostatic network on Sho expression, we investigated the action of lactacystin, MG132, NH4Cl, and 3-methyladenine (3-MA) in two cell culture models. In primary mixed neuronal and glial cell cultures (MNGCs) from transgenic mice expressing wild-type Sho from the PrP gene promoter (Tg.Sprn mice), lactacystin- and MG132-mediated inhibition of proteasomal activity shifted the repertoire of Sho species towards unglycosylated forms appearing in the nuclei; conversely, the autophagic modulators NH4Cl and 3-MA did not affect Sho or PrPC glycosylation patterns. Mouse N2a neuroblastoma cells expressing Sho under control of a housekeeping gene promoter treated with MG132 or lactacystin also showed increased nuclear localization of unglycosylated Sho. As two proteasomal inhibitors tested in two cell paradigms caused redirection of Sho to nuclei at the expense of processing through the secretory pathway, our findings define a balanced shift in subcellular localization that thereby differs from the decreases in net Sho species seen in prion-infected brains. Our data are indicative of a physiological pathway to access Sho functions in the nucleus under conditions of impaired proteasomal activity. We also infer that these conditions would comprise a context wherein Sho's N-terminal nucleic acid-binding RGG repeat region is brought into play.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Priones/metabolismo , Inhibidores de Proteasoma/farmacología , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Proteínas Ligadas a GPI , Humanos , Leupeptinas/farmacología , Ratones Noqueados , Modelos Biológicos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Regiones Promotoras Genéticas/genética
15.
Brain Res ; 1208: 170-80, 2008 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-18395703

RESUMEN

The cerebellum is involved in complex physiological functions including motor control, sensory perception, cognition, language, and emotion. Humans and animals with prion diseases are characterized clinically by ataxia, postural abnormalities and cognitive decline. Pathology in the cerebellum affected by prions includes spongiform degeneration, neuronal loss, and gliosis. To develop an in vitro model system for studying prion biology in cerebellar cells, we established and characterized an immortal cell line (CRBL) isolated from the cerebellum of mice lacking expression of a protein involved in cell cycle arrest. The characteristics of the cells include morphological heterogeneity, rapid proliferation, serum responsiveness during growth, and a change in the number of chromosomes. CRBL cells expressed both neuronal and glial cell markers as well as a considerable level of cellular prion protein, PrP(C). Upon in vitro infection, CRBL cells exhibited selective susceptibility to prions isolated from different sources. These cells chronically propagated prions from SMB cells. Strain-specific prion infection in CRBL cells was not due to instability of the cell line, allelic variance, or mutations in the PrP gene. Molecular properties of prions derived from SMB cells were maintained in the infected CRBL cells. Our results suggest that the specific interaction between a prion strain and hosts determined the selective susceptibility of CRBL cells, which reflects the conditions in vivo. In addition to the future studies revealing cellular and molecular mechanism involved in prion pathogenesis, CRBL cells will contribute to the studies dealing with prion strain properties and host susceptibilities.


Asunto(s)
Línea Celular Transformada/fisiología , Susceptibilidad a Enfermedades , Neuronas/fisiología , Priones/metabolismo , Animales , Recuento de Células , Células Cultivadas , Cerebelo/citología , Citogenética/métodos , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicosilación , Ratones , Ratones Noqueados , Transfección/métodos , Tubulina (Proteína)/metabolismo , Proteína p53 Supresora de Tumor/deficiencia
16.
Int J Med Sci ; 5(6): 347-53, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19015743

RESUMEN

Prion diseases are caused by proteinaceous pathogens termed prions. Although the details of the mechanism of prion propagation are not fully understood, conformational conversion of cellular prion protein (PrP(C)) to misfolded, disease-associated scrapie prion protein (PrP(Sc)) is considered the essential biochemical event for prion replication. Currently, studying prion replication in vitro is difficult due to the lack of a system which fully recapitulates the in vivo phenomenon. Over the last 15 years, a number of in vitro systems supporting PrP(C) conversion, PrP(Sc) amplification, or amyloid fibril formation have been established. In this review, we describe the evolving methodology of in vitro prion propagation assays and discuss their ability in reflecting prion propagation in vivo.


Asunto(s)
Priones/metabolismo , Animales , Biocatálisis , Sistema Libre de Células , Humanos , Priones/genética , Desnaturalización Proteica , Pliegue de Proteína
17.
Brain Res ; 1648(Pt B): 553-560, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27060771

RESUMEN

Prion diseases are fatal neurodegenerative disorders that include scrapie of sheep, bovine spongiform encephalopathy of cattle, chronic wasting disease of cervids, and Creutzfeldt-Jakob disease (CJD) of humans. The etiology for prion diseases can be infectious, sporadic, or hereditary. However, the common denominator for all types is the formation of a transmissible agent composed of a ß-sheet-rich, misfolded version of the host-encoded prion protein (PrPC), known as PrPSc. PrPSc self-replicates through a template-assisted process that converts the α-helical conformation of PrPC into the disease-associated isoform. In parallel with PrPSc accumulation, spongiform change is pathologically observed in the central nervous system, where "holes" appear because of massive neuronal death. Here, we review the cellular pathways triggered in response to PrPSc formation and accumulation. Available data suggest that neuronal dysfunction and death may be caused by what originates as a cellular pro-survival response to chronic PrPSc accumulation. We also discuss what is known about the complex cross-talk between the endoplasmic reticulum stress components and the quality control pathways. Better knowledge about these processes may lead to innovative therapeutic strategies based on manipulating the stress response and its consequences for neurodegeneration. This article is part of a Special Issue entitled SI:ER stress.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Enfermedades por Prión/patología , Enfermedades por Prión/fisiopatología , Animales , Humanos , Proteínas Priónicas/metabolismo , Transducción de Señal/fisiología
18.
Mol Biol Cell ; 27(5): 757-67, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26764098

RESUMEN

Prion diseases are fatal neurodegenerative disorders for which there is no effective treatment. Because the cellular prion protein (PrP(C)) is required for propagation of the infectious scrapie form of the protein, one therapeutic strategy is to reduce PrP(C) expression. Recently FK506, an inhibitor of the FKBP family of peptidyl prolyl isomerases, was shown to increase survival in animal models of prion disease, with proposed mechanisms including calcineurin inhibition, induction of autophagy, and reduced PrP(C) expression. We show that FK506 treatment results in a profound reduction in PrP(C) expression due to a defect in the translocation of PrP(C) into the endoplasmic reticulum with subsequent degradation by the proteasome. These phenotypes could be bypassed by replacing the PrP(C) signal sequence with that of prolactin or osteopontin. In mouse cells, depletion of ER luminal FKBP10 was almost as potent as FK506 in attenuating expression of PrP(C). However, this occurred at a later stage, after translocation of PrP(C) into the ER. Both FK506 treatment and FKBP10 depletion were effective in reducing PrP(Sc) propagation in cell models. These findings show the involvement of FKBP proteins at different stages of PrP(C) biogenesis and identify FKBP10 as a potential therapeutic target for the treatment of prion diseases.


Asunto(s)
Antiinfecciosos/farmacología , Proteínas PrPC/metabolismo , Tacrolimus/farmacología , Animales , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Células Hep G2/efectos de los fármacos , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Transporte de Proteínas/efectos de los fármacos , Scrapie/tratamiento farmacológico , Scrapie/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
19.
EMBO Mol Med ; 7(3): 339-56, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25661904

RESUMEN

The cellular prion protein (PrP(C)) comprises a natively unstructured N-terminal domain, including a metal-binding octarepeat region (OR) and a linker, followed by a C-terminal domain that misfolds to form PrP(S) (c) in Creutzfeldt-Jakob disease. PrP(C) ß-endoproteolysis to the C2 fragment allows PrP(S) (c) formation, while α-endoproteolysis blocks production. To examine the OR, we used structure-directed design to make novel alleles, 'S1' and 'S3', locking this region in extended or compact conformations, respectively. S1 and S3 PrP resembled WT PrP in supporting peripheral nerve myelination. Prion-infected S1 and S3 transgenic mice both accumulated similar low levels of PrP(S) (c) and infectious prion particles, but differed in their clinical presentation. Unexpectedly, S3 PrP overproduced C2 fragment in the brain by a mechanism distinct from metal-catalysed hydrolysis reported previously. OR flexibility is concluded to impact diverse biological endpoints; it is a salient variable in infectious disease paradigms and modulates how the levels of PrP(S) (c) and infectivity can either uncouple or engage to drive the onset of clinical disease.


Asunto(s)
Proteínas PrPC/química , Proteínas PrPC/metabolismo , Enfermedades por Prión/patología , Enfermedades por Prión/fisiopatología , Procesamiento Proteico-Postraduccional , Animales , Línea Celular , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Histocitoquímica , Humanos , Ratones Transgénicos , Microscopía , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformación Proteica , Proteolisis
20.
FEBS J ; 281(3): 862-76, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24286250

RESUMEN

Cellular prion protein (PrP(C)) misfolds to form infectivity-associated scrapie prion protein and generates C-terminal fragments C1 and C2 in healthy and prion-infected animals. C1 cleavage occurs N-terminally of PrP(C)'s hydrophobic domain, whereas the larger C2 fragment is generated by cleavage at the end of the octarepeat region. As the PrP-like proteins Doppel and Shadoo (Sho) have been reported to inhabit similar membrane environments as PrP(C), we investigated endoproteolysis by using a panel of mutant alleles. Doppel undergoes efficient in vivo cleavage at a C1 site mapped to the start of the globular domain, which is a structurally similar cleavage site to that in PrP(C). Sho is processed to C1 and C2 fragments, and proved refractory to mutagenesis to inactivate C1 cleavage. As a reciprocal product of C1 cleavage, Sho also engenders a metabolically stable N1 fragment with a C-terminus after its hydrophobic domain, an observation that may account for N1's association with membrane and/or cellular fractions in vitro and in vivo. Our data indicate that glycosylation status and yet to be identified proteases modulate internal C1 and C2 proteolysis events within the mammalian prion protein family.


Asunto(s)
Endopeptidasas/metabolismo , Glicoproteínas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas PrPC/metabolismo , Priones/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Línea Celular , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Glicosilación , Masculino , Ratones , Ratones Transgénicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/enzimología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas PrPC/química , Proteínas PrPC/genética , Priones/química , Priones/genética , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteolisis , Conejos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Testículo/enzimología , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA