Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Alzheimers Dement ; 20(1): 124-135, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37489764

RESUMO

INTRODUCTION: As the chemokine receptor5 (CCR5) may play a role in ischemia, we studied the links between CCR5 deficiency, the sensitivity of neurons to oxidative stress, and the development of dementia. METHODS: Logistic regression models with CCR5/apolipoprotein E (ApoE) polymorphisms were applied on a sample of 205 cognitively normal individuals and 189 dementia patients from Geneva. The impact of oxidative stress on Ccr5 expression and cell death was assessed in mice neurons. RESULTS: CCR5-Δ32 allele synergized with ApoEε4 as risk factor for dementia and specifically for dementia with a vascular component. We confirmed these results in an independent cohort from Italy (157 cognitively normal and 620 dementia). Carriers of the ApoEε4/CCR5-Δ32 genotype aged ≥80 years have an 11-fold greater risk of vascular-and-mixed dementia. Oxidative stress-induced cell death in Ccr5-/- mice neurons. DISCUSSION: We propose the vulnerability of CCR5-deficient neurons in response to oxidative stress as possible mechanisms contributing to dementia.


Assuntos
Demência Vascular , Resiliência Psicológica , Humanos , Animais , Camundongos , Demência Vascular/genética , Genótipo , Quimiocinas , Polimorfismo Genético , Receptores CCR5/genética
2.
Commun Biol ; 5(1): 557, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35676449

RESUMO

Mammalian models are essential for brain aging research. However, the long lifespan and poor amenability to genetic and pharmacological perturbations have hindered the use of mammals for dissecting aging-regulatory molecular networks and discovering new anti-aging interventions. To circumvent these limitations, we developed an ex vivo model system that faithfully mimics the aging process of the mammalian brain using cultured mouse brain slices. Genome-wide gene expression analyses showed that cultured brain slices spontaneously upregulated senescence-associated genes over time and reproduced many of the transcriptional characteristics of aged brains. Treatment with rapamycin, a classical anti-aging compound, largely abolished the time-dependent transcriptional changes in naturally aged brain slice cultures. Using this model system, we discovered that prions drastically accelerated the development of age-related molecular signatures and the pace of brain aging. We confirmed this finding in mouse models and human victims of Creutzfeldt-Jakob disease. These data establish an innovative, eminently tractable mammalian model of brain aging, and uncover a surprising acceleration of brain aging in prion diseases.


Assuntos
Envelhecimento , Encéfalo , Síndrome de Creutzfeldt-Jakob , Doenças Priônicas , Príons , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Camundongos , Doenças Priônicas/genética , Doenças Priônicas/patologia , Príons/genética
4.
Brain Pathol ; 32(5): e13056, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35178783

RESUMO

Although prion infections cause cognitive impairment and neuronal death, transcriptional and translational profiling shows progressive derangement within glia but surprisingly little changes within neurons. Here we expressed PrPC selectively in neurons and astrocytes of mice. After prion infection, both astrocyte and neuron-restricted PrPC expression led to copious brain accumulation of PrPSc . As expected, neuron-restricted expression was associated with typical prion disease. However, mice with astrocyte-restricted PrPC expression experienced a normal life span, did not develop clinical disease, and did not show astro- or microgliosis. Besides confirming that PrPSc is innocuous to PrPC -deficient neurons, these results show that astrocyte-born PrPSc does not activate the extreme neuroinflammation that accompanies the onset of prion disease and precedes any molecular changes of neurons. This points to a nonautonomous mechanism by which prion-infected neurons instruct astrocytes and microglia to acquire a specific cellular state that, in turn, drives neural dysfunction.


Assuntos
Doenças Priônicas , Príons , Animais , Astrócitos/metabolismo , Camundongos , Neuroglia/metabolismo , Neurônios/metabolismo , Doenças Priônicas/metabolismo , Príons/metabolismo
5.
Nat Struct Mol Biol ; 28(4): 365-372, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33767451

RESUMO

Prions consist of pathological aggregates of cellular prion protein and have the ability to replicate, causing neurodegenerative diseases, a phenomenon mirrored in many other diseases connected to protein aggregation, including Alzheimer's and Parkinson's diseases. However, despite their key importance in disease, the individual processes governing this formation of pathogenic aggregates, as well as their rates, have remained challenging to elucidate in vivo. Here we bring together a mathematical framework with kinetics of the accumulation of prions in mice and microfluidic measurements of aggregate size to dissect the overall aggregation reaction into its constituent processes and quantify the reaction rates in mice. Taken together, the data show that multiplication of prions in vivo is slower than in in vitro experiments, but efficient when compared with other amyloid systems, and displays scaling behavior characteristic of aggregate fragmentation. These results provide a framework for the determination of the mechanisms of disease-associated aggregation processes within living organisms.


Assuntos
Doença de Alzheimer/genética , Doença de Parkinson/genética , Príons/genética , Agregação Patológica de Proteínas/genética , Doença de Alzheimer/patologia , Amiloide/genética , Animais , Humanos , Camundongos , Modelos Teóricos , Doença de Parkinson/patologia
6.
PLoS One ; 15(11): e0242137, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33180885

RESUMO

The adhesion G-protein coupled receptor Adgrg6 (formerly Gpr126) is instrumental in the development, maintenance and repair of peripheral nervous system myelin. The prion protein (PrP) is a potent activator of Adgrg6 and could be used as a potential therapeutic agent in treating peripheral demyelinating and dysmyelinating diseases. We designed a dimeric Fc-fusion protein comprising the myelinotrophic domain of PrP (FT2Fc), which activated Adgrg6 in vitro and exhibited favorable pharmacokinetic properties for in vivo treatment of peripheral neuropathies. While chronic FT2Fc treatment elicited specific transcriptomic changes in the sciatic nerves of PrP knockout mice, no amelioration of the early molecular signs demyelination was detected. Instead, RNA sequencing of sciatic nerves revealed downregulation of cytoskeletal and sarcomere genes, akin to the gene expression changes seen in myopathic skeletal muscle of PrP overexpressing mice. These results call for caution when devising myelinotrophic therapies based on PrP-derived Adgrg6 ligands. While our treatment approach was not successful, Adgrg6 remains an attractive therapeutic target to be addressed in other disease models or by using different biologically active Adgrg6 ligands.


Assuntos
Doenças Desmielinizantes/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Proteínas Priônicas/química , Receptores Acoplados a Proteínas G/agonistas , Animais , Linhagem Celular , Doenças Desmielinizantes/genética , Feminino , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Proteínas Priônicas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Nervo Isquiático/metabolismo , Transcriptoma
7.
EMBO Mol Med ; 12(9): e12739, 2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32776637

RESUMO

Prion immunotherapy may hold great potential, but antibodies against certain PrP epitopes can be neurotoxic. Here, we identified > 6,000 PrP-binding antibodies in a synthetic human Fab phage display library, 49 of which we characterized in detail. Antibodies directed against the flexible tail of PrP conferred neuroprotection against infectious prions. We then mined published repertoires of circulating B cells from healthy humans and found antibodies similar to the protective phage-derived antibodies. When expressed recombinantly, these antibodies exhibited anti-PrP reactivity. Furthermore, we surveyed 48,718 samples from 37,894 hospital patients for the presence of anti-PrP IgGs and found 21 high-titer individuals. The clinical files of these individuals did not reveal any enrichment of specific pathologies, suggesting that anti-PrP autoimmunity is innocuous. The existence of anti-prion antibodies in unbiased human immunological repertoires suggests that they might clear nascent prions early in life. Combined with the reported lack of such antibodies in carriers of disease-associated PRNP mutations, this suggests a link to the low incidence of spontaneous prion diseases in human populations.


Assuntos
Doenças Priônicas , Príons , Anticorpos , Linfócitos B , Humanos , Imunoterapia
8.
Life Sci Alliance ; 3(8)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32576602

RESUMO

HIV and EBV are human pathogens that cause a considerable burden to worldwide health. In combination, these viruses are linked to AIDS-associated lymphomas. We found that EBV, which transforms B cells, renders them susceptible to HIV-1 infection in a CXCR4 and CD4-dependent manner in vitro and that CXCR4-tropic HIV-1 integrates into the genome of these B cells with the same molecular profile as in autologous CD4+ T cells. In addition, we established a humanized mouse model to investigate the in vivo interactions of EBV and HIV-1 upon coinfection. The respective mice that reconstitute human immune system components upon transplantation with CD34+ human hematopoietic progenitor cells could recapitulate aspects of EBV and HIV immunobiology observed in dual-infected patients. Upon coinfection of humanized mice, EBV/HIV dual-infected B cells could be detected, but were susceptible to CD8+ T-cell-mediated immune control.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/virologia , Herpesvirus Humano 4/patogenicidade , Animais , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos B/virologia , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Coinfecção , Modelos Animais de Doenças , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/virologia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por HIV/genética , Soropositividade para HIV , HIV-1/metabolismo , HIV-1/patogenicidade , Células-Tronco Hematopoéticas/patologia , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiologia , Linfócitos T/imunologia
9.
PLoS Pathog ; 16(6): e1008653, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32598380

RESUMO

The clinical course of prion diseases is accurately predictable despite long latency periods, suggesting that prion pathogenesis is driven by precisely timed molecular events. We constructed a searchable genome-wide atlas of mRNA abundance and splicing alterations during the course of disease in prion-inoculated mice. Prion infection induced PrP-dependent transient changes in mRNA abundance and processing already at eight weeks post inoculation, well ahead of any neuropathological and clinical signs. In contrast, microglia-enriched genes displayed an increase simultaneous with the appearance of clinical signs, whereas neuronal-enriched transcripts remained unchanged until the very terminal stage of disease. This suggests that glial pathophysiology, rather than neuronal demise, could be the final driver of disease. The administration of young plasma attenuated the occurrence of early mRNA abundance alterations and delayed signs in the terminal phase of the disease. The early onset of prion-induced molecular changes might thus point to novel biomarkers and potential interventional targets.


Assuntos
Estudo de Associação Genômica Ampla , Microglia/metabolismo , Neurônios/metabolismo , Doenças Priônicas , RNA Mensageiro , Transcriptoma , Animais , Masculino , Camundongos , Camundongos Knockout , Doenças Priônicas/genética , Doenças Priônicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Sci Rep ; 8(1): 14600, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30279468

RESUMO

Prion diseases, Alzheimer's disease and Parkinson's disease (PD) are fatal degenerative disorders that share common neuropathological and biochemical features, including the aggregation of pathological protein conformers. Lymphocyte activation gene 3 (Lag3, also known as CD223) is a member of the immunoglobulin superfamily of receptors expressed on peripheral immune cells, microglia and neurons, which serves as a receptor for α-synuclein aggregates in PD. Here we examined the possible role of Lag3 in the pathogenesis of prion diseases. Through quantitative real-time PCR and RNA-sequencing, we found that the expression levels of Lag3 were relatively low in the adult mouse brains, yet its expression was increased after prion infection. However, we failed finding significant differences regarding the incubation time, PrPSc load, neurodegeneration, astrocyte and microglia reactions and inflammatory gene expression between the Lag3 knockout mice and wild-type littermate controls after prion infection. We conclude that loss of Lag3 has no significant influence on prion disease pathogenesis. Considering that Lag3 is an immune checkpoint receptor, our results suggest that immune checkpoint inhibition (an increasingly prevalent therapeutic modality against many types of cancer) might not exert positive or negative effects on the progression of prion diseases.


Assuntos
Antígenos CD/genética , Encéfalo/patologia , Proteínas PrPSc/genética , Scrapie/genética , Animais , Antígenos CD/imunologia , Astrócitos/imunologia , Astrócitos/patologia , Encéfalo/imunologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Progressão da Doença , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Linfócitos/imunologia , Linfócitos/patologia , Camundongos , Camundongos Knockout , Microglia/imunologia , Microglia/patologia , Neurônios/imunologia , Neurônios/patologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Proteínas PrPSc/imunologia , Proteínas PrPSc/patogenicidade , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Scrapie/imunologia , Scrapie/mortalidade , Scrapie/patologia , Transdução de Sinais , Análise de Sobrevida , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
11.
PLoS Pathog ; 13(11): e1006733, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29176838

RESUMO

Prion infections cause inexorable, progressive neurological dysfunction and neurodegeneration. Expression of the cellular prion protein PrPC is required for toxicity, suggesting the existence of deleterious PrPC-dependent signaling cascades. Because group-I metabotropic glutamate receptors (mGluR1 and mGluR5) can form complexes with the cellular prion protein (PrPC), we investigated the impact of mGluR1 and mGluR5 inhibition on prion toxicity ex vivo and in vivo. We found that pharmacological inhibition of mGluR1 and mGluR5 antagonized dose-dependently the neurotoxicity triggered by prion infection and by prion-mimetic anti-PrPC antibodies in organotypic brain slices. Prion-mimetic antibodies increased mGluR5 clustering around dendritic spines, mimicking the toxicity of Aß oligomers. Oral treatment with the mGluR5 inhibitor, MPEP, delayed the onset of motor deficits and moderately prolonged survival of prion-infected mice. Although group-I mGluR inhibition was not curative, these results suggest that it may alleviate the neurological dysfunctions induced by prion diseases.


Assuntos
Proteínas PrPC/toxicidade , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Piridinas/administração & dosagem , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Animais , Anticorpos/administração & dosagem , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Doenças Priônicas/genética , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo
12.
Free Radic Biol Med ; 112: 387-396, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28811143

RESUMO

Neurodegenerative disease are frequently characterized by microglia activation and/or leukocyte infiltration in the parenchyma of the central nervous system and at the molecular level by increased oxidative modifications of proteins, lipids and nucleic acids. NADPH oxidases (NOX) emerged as a novel promising class of pharmacological targets for the treatment of neurodegeneration due to their role in oxidant generation and presumably in regulating microglia activation. The unique function of NOX is the generation of superoxide anion (O2•-) and hydrogen peroxide (H2O2). However in the context of neuroinflammation, they present paradoxical features since O2•-/H2O2 generated by NOX and/or secondary reactive oxygen species (ROS) derived from O2•-/H2O2 can either lead to neuronal oxidative damage or resolution of inflammation. The role of NOX enzymes has been investigated in many models of neurodegenerative diseases by using either genetic or pharmacological approaches. In the present review we provide a critical assessment of recent findings related to the role of NOX in the CNS as well as how the field has advanced over the last 5 years. In particular, we focus on the data derived from the work of a consortium (Neurinox) funded by the European Commission's Programme 7 (FP7). We discuss the evidence gathered from animal models and human samples linking NOX expression/activity with neuroinflammation in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and Creutzfeldt-Jakob disease as well as autoimmune demyelinating diseases like multiple sclerosis (MS) and chronic inflammatory demyelinating polyneuropathy (CIDP). We address the possibility to use measurement of the activity of the NOX2 isoform in blood samples as biomarker of disease severity and treatment efficacy in neurodegenerative disease. Finally we clarify key controversial aspects in the field of NOX, such as NOX cellular expression in the brain, measurement of NOX activity, impact of genetic deletion of NOX in animal models of neurodegeneration and specificity of NOX inhibitors.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Síndrome de Creutzfeldt-Jakob/enzimologia , Esclerose Múltipla/enzimologia , NADPH Oxidase 2/genética , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/enzimologia , Esclerose Lateral Amiotrófica/diagnóstico , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/patologia , Animais , Antioxidantes/uso terapêutico , Biomarcadores/sangue , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/enzimologia , Sistema Nervoso Central/patologia , Síndrome de Creutzfeldt-Jakob/diagnóstico , Síndrome de Creutzfeldt-Jakob/tratamento farmacológico , Síndrome de Creutzfeldt-Jakob/patologia , Modelos Animais de Doenças , Europa (Continente) , Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Cooperação Internacional , Microglia/efeitos dos fármacos , Microglia/enzimologia , Microglia/patologia , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , NADPH Oxidase 2/antagonistas & inibidores , NADPH Oxidase 2/sangue , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/diagnóstico , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/tratamento farmacológico , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/patologia , Superóxidos/metabolismo
13.
PLoS One ; 12(5): e0177876, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28545141

RESUMO

Prion diseases are neurodegenerative conditions caused by misfolding of the prion protein, leading to conspicuous neuronal loss and intense microgliosis. Recent experimental evidence point towards a protective role of microglia against prion-induced neurodegeneration, possibly through elimination of prion-containing apoptotic bodies. The molecular mechanisms by which microglia recognize and eliminate apoptotic cells in the context of prion diseases are poorly defined. Here we investigated the possible involvement of signal regulatory protein α (SIRPα), a key modulator of host cell phagocytosis; SIRPα is encoded by the Sirpa gene that is genetically linked to the prion gene Prnp. We found that Sirpa transcripts are highly enriched in microglia cells within the brain. However, Sirpa mRNA levels were essentially unaltered during the course of experimental prion disease despite upregulation of other microglia-enriched transcripts. To study the involvement of SIRPα in prion pathogenesis in vivo, mice expressing a truncated SIRPα protein unable to inhibit phagocytosis were inoculated with rodent-adapted scrapie prions of the 22L strain. Homozygous and heterozygous Sirpa mutants and wild-type mice experienced similar incubation times after inoculation with either of two doses of 22L prions. Moreover, the extent of neuronal loss, microgliosis and abnormal prion protein accumulation was not significantly affected by Sirpa genotypes. Collectively, these data indicate that SIRPα-mediated phagocytosis is not a major determinant in prion disease pathogenesis. It will be important to search for additional candidates mediating prion phagocytosis, as this mechanism may represent an important target of antiprion therapies.


Assuntos
Microglia/metabolismo , Doenças Priônicas/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Animais , Progressão da Doença , Regulação da Expressão Gênica , Camundongos , Microglia/patologia , Mutação , Fagocitose , Doenças Priônicas/genética , Doenças Priônicas/patologia , Proteínas Priônicas/metabolismo
16.
PLoS One ; 12(2): e0171923, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28178353

RESUMO

Misfolding of the cellular prion protein (PrPC) into the scrapie prion protein (PrPSc) results in progressive, fatal, transmissible neurodegenerative conditions termed prion diseases. Experimental and epidemiological evidence point toward a protracted, clinically silent phase in prion diseases, yet there is no diagnostic test capable of identifying asymptomatic individuals incubating prions. In an effort to identify early biomarkers of prion diseases, we have compared global transcriptional profiles in brains from pre-symptomatic prion-infected mice and controls. We identified Cst7, which encodes cystatin F, as the most strongly upregulated transcript in this model. Early and robust upregulation of Cst7 mRNA levels and of its cognate protein was validated in additional mouse models of prion disease. Surprisingly, we found no significant increase in cystatin F levels in both cerebrospinal fluid or brain parenchyma of patients with Creutzfeldt-Jakob disease compared to Alzheimer's disease or non-demented controls. Our results validate cystatin F as a useful biomarker of early pathogenesis in experimental models of prion disease, and point to unexpected species-specific differences in the transcriptional responses to prion infections.


Assuntos
Cistatinas/metabolismo , Doenças Priônicas/metabolismo , Animais , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patologia , Cistatinas/líquido cefalorraquidiano , Cistatinas/genética , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Doenças Priônicas/líquido cefalorraquidiano , Doenças Priônicas/genética , Doenças Priônicas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
17.
PLoS One ; 11(9): e0163601, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27684562

RESUMO

Prions are the infectious agents causing transmissible spongiform encephalopathies (TSE), progressive, inexorably lethal neurological diseases. Antibodies targeting the globular domain (GD) of the cellular prion protein PrPC trigger a neurotoxic syndrome morphologically and molecularly similar to prion disease. This phenomenon raises the question whether such antibodies induce infectious prions de novo. Here we exposed cerebellar organotypic cultured slices (COCS) to the neurotoxic antibody, POM1. We then inoculated COCS homogenates into tga20 mice, which overexpress PrPC and are commonly utilized as sensitive indicators of prion infectivity. None of the mice inoculated with COCS-derived lysates developed any signs of disease, and all mice survived for at least 200 days post-inoculation. In contrast, all mice inoculated with bona fide prions succumbed to TSE after 55-95 days. Post-mortem analyses did not reveal any signs of prion pathology in mice inoculated with POM1-COCS lysates. Also, lysates from POM1-exposed COCS were unable to convert PrP by quaking. Hence, anti-GD antibodies do not catalyze the generation of prion infectivity. These data indicate that prion replication can be separated from prion toxicity, and suggest that anti-GD antibodies exert toxicity by acting downstream of prion replication.

18.
Free Radic Biol Med ; 97: 95-108, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27212019

RESUMO

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease characterized by progressive loss of motor neurons, gliosis, neuroinflammation and oxidative stress. The aim of this study was to evaluate the involvement of NADPH oxidases (NOX) in the oxidative damage and progression of ALS neuropathology. We examined the pattern of NOX expression in spinal cords of patients and mouse models of ALS and analyzed the impact of genetic deletion of the NOX1 and 2 isoforms as well as pharmacological NOX inhibition in the SOD1(G93A) ALS mouse model. A substantial (10-60 times) increase of NOX2 expression was detected in three etiologically different ALS mouse models while up-regulation of some other NOX isoforms was model-specific. In human spinal cord samples, high NOX2 expression was detected in microglia. In contrast to previous publications, survival of SOD1(G93A) mice was not modified upon breeding with constitutive NOX1 and NOX2 deficient mice. As genetic deficiency of a single NOX isoform is not necessarily predictive of a pharmacological intervention, we treated SOD1(G93A) mice with broad-spectrum NOX inhibitors perphenazine and thioridazine. Both compounds reached in vivo CNS concentrations compatible with NOX inhibition and thioridazine significantly decreased superoxide levels in the spinal cord of SOD1(G93A) mice in vivo. Yet, neither perphenazine nor thioridazine prolonged survival. Thioridazine, but not perphenazine, dampened the increase of microglia markers in SOD1(G93A) mice. Thioridazine induced an immediate and temporary enhancement of motor performance (rotarod) but its precise mode of action needs further investigation. Additional studies using specific NOX inhibitors will provide further evidence on the relevance of NOX as drug targets for ALS and other neurodegenerative disorders.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , NADPH Oxidase 1/genética , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Adulto , Idoso , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Perfenazina/administração & dosagem , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Superóxido Dismutase-1/antagonistas & inibidores , Superóxido Dismutase-1/genética , Tioridazina/administração & dosagem
19.
J Exp Med ; 213(3): 313-27, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26926995

RESUMO

Although its involvement in prion replication and neurotoxicity during transmissible spongiform encephalopathies is undisputed, the physiological role of the cellular prion protein (PrP(C)) remains enigmatic. A plethora of functions have been ascribed to PrP(C) based on phenotypes of Prnp(-/-) mice. However, all currently available Prnp(-/-) lines were generated in embryonic stem cells from the 129 strain of the laboratory mouse and mostly crossed to non-129 strains. Therefore, Prnp-linked loci polymorphic between 129 and the backcrossing strain resulted in systematic genetic confounders and led to erroneous conclusions. We used TALEN-mediated genome editing in fertilized mouse oocytes to create the Zurich-3 (ZH3) Prnp-ablated allele on a pure C57BL/6J genetic background. Genomic, transcriptional, and phenotypic characterization of Prnp(ZH3/ZH3) mice failed to identify phenotypes previously described in non-co-isogenic Prnp(-/-) mice. However, aged Prnp(ZH3/ZH3) mice developed a chronic demyelinating peripheral neuropathy, confirming the crucial involvement of PrP(C) in peripheral myelin maintenance. This new line represents a rigorous genetic resource for studying the role of PrP(C) in physiology and disease.


Assuntos
Príons/metabolismo , Animais , Sequência de Bases , Cromossomos de Mamíferos , Endonucleases/metabolismo , Feminino , Deleção de Genes , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Degeneração Neural/patologia , Fases de Leitura Aberta/genética , Fagocitose , Fenótipo , Polirradiculoneuropatia/patologia , RNA/metabolismo , Análise de Sequência de RNA , Transativadores/genética
20.
PLoS Pathog ; 11(2): e1004662, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25710374

RESUMO

Prions induce lethal neurodegeneration and consist of PrPSc, an aggregated conformer of the cellular prion protein PrPC. Antibody-derived ligands to the globular domain of PrPC (collectively termed GDL) are also neurotoxic. Here we show that GDL and prion infections activate the same pathways. Firstly, both GDL and prion infection of cerebellar organotypic cultured slices (COCS) induced the production of reactive oxygen species (ROS). Accordingly, ROS scavenging, which counteracts GDL toxicity in vitro and in vivo, prolonged the lifespan of prion-infected mice and protected prion-infected COCS from neurodegeneration. Instead, neither glutamate receptor antagonists nor inhibitors of endoplasmic reticulum calcium channels abolished neurotoxicity in either model. Secondly, antibodies against the flexible tail (FT) of PrPC reduced neurotoxicity in both GDL-exposed and prion-infected COCS, suggesting that the FT executes toxicity in both paradigms. Thirdly, the PERK pathway of the unfolded protein response was activated in both models. Finally, 80% of transcriptionally downregulated genes overlapped between prion-infected and GDL-treated COCS. We conclude that GDL mimic the interaction of PrPSc with PrPC, thereby triggering the downstream events characteristic of prion infection.


Assuntos
Anticorpos , Proteínas PrPSc/imunologia , Doenças Priônicas/induzido quimicamente , Doenças Priônicas/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Animais , Anticorpos/imunologia , Anticorpos/toxicidade , Camundongos , Camundongos Transgênicos , Proteínas PrPSc/genética , Doenças Priônicas/genética , Doenças Priônicas/patologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais/genética , eIF-2 Quinase/genética , eIF-2 Quinase/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...