Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Mol Neurosci ; 15: 990136, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36117913

RESUMO

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.

2.
J Biol Chem ; 294(37): 13619-13628, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31320473

RESUMO

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.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Doenças Priônicas/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Progressão da Doença , Retículo Endoplasmático/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Feminino , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Priônicas/metabolismo , Príons/metabolismo , Dobramento de Proteína
3.
Mol Neurobiol ; 56(11): 7888-7904, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31129810

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Príons/metabolismo , Inibidores de Proteassoma/farmacologia , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Proteínas Ligadas por GPI , Humanos , Leupeptinas/farmacologia , Camundongos Knockout , Modelos Biológicos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética
4.
Sci Rep ; 9(1): 4847, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30890734

RESUMO

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.


Assuntos
Produtos Biológicos/farmacologia , Biotecnologia/métodos , Síndrome de Creutzfeldt-Jakob/tratamento farmacológico , Príons/efeitos dos fármacos , Animais , Linhagem Celular , Humanos , Dobramento de Proteína/efeitos dos fármacos , Coelhos , Sensibilidade e Especificidade
5.
Mol Psychiatry ; 23(12): 2363-2374, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30171212

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Organoides/metabolismo , Proteínas tau/metabolismo , Idoso , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/fisiologia , Encéfalo/metabolismo , Técnicas de Cultura de Células/métodos , Córtex Cerebral , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Masculino , Pessoa de Meia-Idade , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Fosforilação , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/fisiologia
6.
Cell Mol Life Sci ; 75(19): 3521-3538, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30030591

RESUMO

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.


Assuntos
Produtos Biológicos/farmacologia , Fenóis/farmacologia , Agregação Patológica de Proteínas/prevenção & controle , Dobramento de Proteína/efeitos dos fármacos , Deficiências na Proteostase/prevenção & controle , Amiloidose/tratamento farmacológico , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Produtos Biológicos/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Humanos , Fenóis/uso terapêutico , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Deficiências na Proteostase/tratamento farmacológico
7.
PLoS Pathog ; 14(1): e1006826, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29338055

RESUMO

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.


Assuntos
Doença de Gerstmann-Straussler-Scheinker/genética , Mutação , Proteínas PrPSc/química , Proteínas PrPSc/genética , Doenças Priônicas/genética , Adulto , Alelos , Sequência de Aminoácidos , Animais , Humanos , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/genética , Proteínas PrPSc/metabolismo , Domínios Proteicos/genética , Precursores de Proteínas/química , Precursores de Proteínas/genética
8.
Brain Res ; 1648(Pt B): 553-560, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27060771

RESUMO

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.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Doenças Priônicas/patologia , Doenças Priônicas/fisiopatologia , Animais , Humanos , Proteínas Priônicas/metabolismo , Transdução de Sinais/fisiologia
9.
Mol Biol Cell ; 27(5): 757-67, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26764098

RESUMO

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.


Assuntos
Anti-Infecciosos/farmacologia , Proteínas PrPC/metabolismo , Tacrolimo/farmacologia , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Células Hep G2/efeitos dos fármacos , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico/efeitos dos fármacos , Scrapie/tratamento farmacológico , Scrapie/metabolismo , Proteínas de Ligação a Tacrolimo/antagonistas & inibidores , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo
10.
J Virol ; 89(24): 12418-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26423957

RESUMO

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).


Assuntos
Encéfalo/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/metabolismo , Dobramento de Proteína , Multimerização Proteica , Animais , Encéfalo/patologia , Camundongos , Camundongos Transgênicos , Proteínas PrPSc/genética , Doenças Priônicas/genética , Doenças Priônicas/patologia
11.
EMBO Mol Med ; 7(3): 339-56, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25661904

RESUMO

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.


Assuntos
Proteínas PrPC/química , Proteínas PrPC/metabolismo , Doenças Priônicas/patologia , Doenças Priônicas/fisiopatologia , Processamento de Proteína Pós-Traducional , Animais , Linhagem Celular , Análise Mutacional de DNA , Modelos Animais de Doenças , Histocitoquímica , Humanos , Camundongos Transgênicos , Microscopia , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformação Proteica , Proteólise
12.
J Clin Invest ; 124(2): 847-58, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24430187

RESUMO

The symptoms of prion infection can take years or decades to manifest following the initial exposure. Molecular markers of prion disease include accumulation of the misfolded prion protein (PrPSc), which is derived from its cellular precursor (PrPC), as well as downregulation of the PrP-like Shadoo (Sho) glycoprotein. Given the overlapping cellular environments for PrPC and Sho, we inferred that PrPC levels might also be altered as part of a host response during prion infection. Using rodent models, we found that, in addition to changes in PrPC glycosylation and proteolytic processing, net reductions in PrPC occur in a wide range of prion diseases, including sheep scrapie, human Creutzfeldt-Jakob disease, and cervid chronic wasting disease. The reduction in PrPC results in decreased prion replication, as measured by the protein misfolding cyclic amplification technique for generating PrPSc in vitro. While PrPC downregulation is not discernible in animals with unusually short incubation periods and high PrPC expression, slowly evolving prion infections exhibit downregulation of the PrPC substrate required for new PrPSc synthesis and as a receptor for pathogenic signaling. Our data reveal PrPC downregulation as a previously unappreciated element of disease pathogenesis that defines the extensive, presymptomatic period for many prion strains.


Assuntos
Doenças Priônicas/fisiopatologia , Animais , Arvicolinae , Encéfalo/metabolismo , Linhagem Celular , Síndrome de Creutzfeldt-Jakob/metabolismo , Síndrome de Creutzfeldt-Jakob/fisiopatologia , Progressão da Doença , Regulação para Baixo , Glicosilação , Humanos , Mesocricetus , Camundongos , Camundongos Transgênicos , Proteínas PrPC/química , Proteínas PrPSc/química , Doenças Priônicas/metabolismo , Isoformas de Proteínas/química , Scrapie/metabolismo , Scrapie/fisiopatologia , Transdução de Sinais , Fatores de Tempo , Doença de Emaciação Crônica
13.
FEBS J ; 281(3): 862-76, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24286250

RESUMO

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.


Assuntos
Endopeptidases/metabolismo , Glicoproteínas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Príons/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Linhagem Celular , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Glicosilação , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/enzimologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas PrPC/química , Proteínas PrPC/genética , Príons/química , Príons/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteólise , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Testículo/enzimologia , Testículo/metabolismo
14.
PLoS Pathog ; 9(11): e1003755, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244171

RESUMO

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.


Assuntos
Proliferação de Células , Proteínas PrPSc/metabolismo , Animais , Linhagem Celular , Cricetinae , Camundongos , Fibras Musculares Esqueléticas , Especificidade da Espécie
15.
Biochemistry ; 51(45): 9029-31, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23121093

RESUMO

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.


Assuntos
Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , RNA Bacteriano/metabolismo , Animais , Arginina/química , Proteínas Ligadas por GPI , Lisina/química , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Repetições de Trinucleotídeos/fisiologia
16.
Biomaterials ; 33(28): 6808-22, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22748770

RESUMO

Quinacrine and related heterocyclic compounds have antiprion activity. Since the infectious pathogen of prion diseases is composed of multimeric PrP(Sc) assemblies, we hypothesized that this antiprion property could be enhanced by attaching multiple quinacrine-derived chloroquinoline or acridine moieties to a scaffold. In addition to exploring Congo red dye and tetraphenylporphyrin tetracarboxylic acid scaffolds, which already possess intrinsic prion-binding ability; trimesic acid was used in this role. In practice, Congo red itself could not be modified with chloroquinoline or acridine units, and a modified dicarboxyl analog was also unreactive. The latter also lacked antiprion activity in infected cultured cells. While addition of chloroquinoline to a tetraphenylporphyrin tetracarboxylic acid scaffold resulted in some reduction of PrP(Sc), moieties attached to a trimesic acid scaffold exhibited sub-micromolar IC(50)'s as well as a toxicity profile superior to quinacrine. Antiprion activity of these molecules was influenced by the length, polarity, and rigidity associated with the variable linear or cyclic polyamine tethers, and in some instances was modulated by host-cell and/or strain type. Unexpectedly, several compounds in our series increased PrP(Sc) levels. Overall, inhibitory and enhancing properties of these multivalent compounds offer new avenues for structure-based investigation of prion biology.


Assuntos
Doenças Priônicas/tratamento farmacológico , Príons/antagonistas & inibidores , Quinacrina/química , Quinacrina/farmacologia , Ácidos Tricarboxílicos/química , Ácidos Tricarboxílicos/farmacologia , Animais , Linhagem Celular Tumoral/efeitos dos fármacos , Cloroquinolinóis/química , Cloroquinolinóis/farmacologia , Vermelho Congo/química , Vermelho Congo/farmacologia , Humanos , Concentração Inibidora 50 , Camundongos , Porfirinas/química , Porfirinas/farmacologia , Proteínas PrPSc/antagonistas & inibidores , Relação Estrutura-Atividade
17.
PLoS Pathog ; 7(11): e1002391, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22114562

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas PrPSc/biossíntese , Doenças Priônicas/metabolismo , Animais , Regulação para Baixo , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas PrPC/metabolismo , RNA Mensageiro/metabolismo
18.
PLoS One ; 6(3): e18047, 2011 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-21464935

RESUMO

Protein misfolding cyclic amplification (PMCA) recapitulates the prion protein (PrP) conversion process under cell-free conditions. PMCA was initially established with brain material and then with further simplified constituents such as partially purified and recombinant PrP. However, availability of brain material from some species or brain material from animals with certain mutations or polymorphisms within the PrP gene is often limited. Moreover, preparation of native PrP from mammalian cells and tissues, as well as recombinant PrP from bacterial cells, involves time-consuming purification steps. To establish a convenient and versatile PMCA procedure unrestricted to the availability of substrate sources, we attempted to conduct PMCA with the lysate of cells that express cellular PrP (PrP(C)). PrP(Sc) was efficiently amplified with lysate of rabbit kidney epithelial RK13 cells stably transfected with the mouse or Syrian hamster PrP gene. Furthermore, PMCA was also successful with lysate of other established cell lines of neuronal or non-neuronal origins. Together with the data showing that the abundance of PrP(C) in cell lysate was a critical factor to drive efficient PrP(Sc) amplification, our results demonstrate that cell lysate in which PrP(C) is present abundantly serves as an excellent substrate source for PMCA.


Assuntos
Bioquímica/métodos , Extratos Celulares/análise , Príons/metabolismo , Dobramento de Proteína , Animais , Células Cultivadas , Cricetinae , Camundongos , Neurônios/metabolismo , Fases de Leitura Aberta/genética , Proteínas PrPSc/metabolismo , Príons/genética , Coelhos
19.
Biomaterials ; 32(11): 3141-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21288569

RESUMO

Poly-l-lysine (PLL), a homopolymer of amino acid l-lysine (LL), has been frequently used for drug delivery. Here, we report that PLL is an effective agent to inhibit propagation of prions that cause fatal and incurable neurologic disorders in humans and animals, termed prion diseases. In our recent investigation on prion propagation facilitated by conversion of the cellular prion protein (PrP) to the misfolded, disease-associated PrP (PrP(Sc)), we demonstrated that plasminogen stimulates PrP conversion as a cellular cofactor. In the current study, we targeted plasminogen using PLL and assessed its anti-prion efficacy. The results showed that PLL strongly inhibited PrP(Sc) propagation in the cell-free, cell culture, and mouse models of prion disease. These results confirm the role of plasminogen in PrP(Sc) propagation, validates plasminogen as a therapeutic target to combat prion disease, and suggests PLL as a potential anti-prion agent. Therefore, our study represents a proof-of-concept that targeting plasminogen, a cofactor for PrP conversion, using PLL results in suppression of prion propagation, which represents a successful translation of our understanding on details of prion propagation into a potential therapeutic strategy for prion diseases.


Assuntos
Plasminogênio/metabolismo , Polilisina/química , Polilisina/uso terapêutico , Doenças Priônicas/tratamento farmacológico , Príons/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Polilisina/administração & dosagem , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/metabolismo
20.
Prion ; 5(1): 22-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21191173

RESUMO

The biochemical essence of prion replication is the molecular multiplication of the disease-associated misfolded isoform of prion protein (PrP), termed PrPSc, in a nucleic acid-free manner. PrP(Sc) is generated by the protein misfolding process facilitated by conformational conversion of the host-encoded cellular PrP to PrP(Sc). Evidence suggests that an auxiliary factor may play a role in PrP(Sc) propagation. We and others previously discovered that plasminogen interacts with PrP, while its functional role for PrPSc propagation remained undetermined. In our recent in vitro PrP conversion study, we showed that plasminogen substantially stimulates PrP(Sc) propagation in a concentration-dependent manner by accelerating the rate of PrP(Sc) generation, while depletion of plasminogen, destabilization of its structure, and interference with the PrP-plasminogen interaction hinder PrP(Sc) propagation. Further investigation in cell culture models confirmed an increase of PrP(Sc) formation by plasminogen. Although molecular basis of the observed activity for plasminogen remain to be addressed, our results demonstrate that plasminogen is the first cellular protein auxiliary factor proven to stimulate PrP(Sc) propagation.


Assuntos
Plasminogênio/metabolismo , Proteínas PrPSc/metabolismo , Animais , Humanos , Peptídeos/química , Peptídeos/metabolismo , Plasminogênio/química , Proteínas PrPSc/química , Dobramento de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...