Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nat Sci Sleep ; 15: 677-690, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37621720

RESUMO

Purpose: Differential diagnosis of central disorders of hypersomnolence remains challenging, particularly between idiopathic (IH) and nonorganic hypersomnia (NOH). We hypothesized that eyelid closure behavior in the maintenance of wakefulness test (MWT) could be a valuable biomarker. Patients and Methods: MWT recordings of patients with IH, NOH, narcolepsy-cataplexy (NC), and healthy sleep-deprived controls (H) were retrospectively analyzed (15 individuals per group). For each MWT trial, visual scoring of face videography for partial (50-80%) and full eyelid closure (≥80%) was performed from "lights off" to the first microsleep episode (≥3 s). Results: In all groups, the frequency and cumulative duration of periods with partial and full eyelid closure gradually increased toward the first microsleep episode. On the group level, significant differences occurred for the latency to the first microsleep episode (IH 21 min (18-33), NOH 23 min (17-35), NC 11 min (7-19), H 10 min (6-25); p = 0.009), the ratio between partial and full eyelid closure duration (IH 2.2 (0.9-3.1), NOH 0.5 (0-1.2), NC 2.8 (1.1-5), H 0.7 (0.4-3.3); p = 0.004), and the difference between full and partial eyelid closure duration in the five minutes prior to the first microsleep episode (∆full - partial eyelid closure duration: IH -16 s (-35 to 28); NOH 46 s (9-82); NC -6 s (-26 to 5); H 10 s (-4 to 18); p = 0.007). IH and NOH significantly differed comparing the ratio between partial and full eyelid closure (p = 0.005) and the difference between ∆full - partial eyelid closure duration in the five minutes prior to the first microsleep episode (p = 0.006). Conclusion: In the MWT, eyelid closure behavior (∆full - partial) in the period prior to the first microsleep episode could be of value for discriminating NOH from other etiologies of excessive daytime sleepiness, particularly IH.

3.
Brain Behav Immun ; 114: 511-522, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37369340

RESUMO

Among systemic lupus erythematosus (SLE) patients, neuropsychiatric symptoms are highly prevalent, being observed in up to 80% of adult and 95% of pediatric patients. Type 1 interferons, particularly interferon alpha (IFNα), have been implicated in the pathogenesis of SLE and its associated neuropsychiatric symptoms (NPSLE). However, it remains unclear how type 1 interferon signaling in the central nervous system (CNS) might result in neuropsychiatric sequelae. In this study, we validate an NPSLE mouse model and find an elevated peripheral type 1 interferon signature alongside clinically relevant NPSLE symptoms such as anxiety and fatigue. Unbiased single-nucleus sequencing of the hindbrain and hippocampus revealed that interferon-stimulated genes (ISGs) were among the most highly upregulated genes in both regions and that gene pathways involved in cellular interaction and neuronal development were generally repressed among astrocytes, oligodendrocytes, and neurons. Using image-based spatial transcriptomics, we found that the type 1 interferon signature is enriched as spatially distinct patches within the brain parenchyma of these mice. Our results suggest that type 1 interferon in the CNS may play an important mechanistic role in mediating NPSLE behavioral phenotypes by repressing general cellular communication pathways, and that type 1 interferon signaling modulators are a potential therapeutic option for NPSLE.


Assuntos
Lúpus Eritematoso Sistêmico , Vasculite Associada ao Lúpus do Sistema Nervoso Central , Adulto , Humanos , Criança , Animais , Camundongos , Vasculite Associada ao Lúpus do Sistema Nervoso Central/complicações , Vasculite Associada ao Lúpus do Sistema Nervoso Central/diagnóstico , Vasculite Associada ao Lúpus do Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Lúpus Eritematoso Sistêmico/complicações , Encéfalo/metabolismo , Interferon-alfa/metabolismo
4.
bioRxiv ; 2023 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-37131759

RESUMO

bioRxiv has withdrawn this manuscript because it was posted without the consent of all authors. If you have any questions, please contact the corresponding author.

5.
Adv Immunol ; 152: 83-155, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34844710

RESUMO

Neuropsychiatric diseases have traditionally been studied from brain, and mind-centric perspectives. However, mounting epidemiological and clinical evidence shows a strong correlation of neuropsychiatric manifestations with immune system activation, suggesting a likely mechanistic interaction between the immune and nervous systems in mediating neuropsychiatric disease. Indeed, immune mediators such as cytokines, antibodies, and complement proteins have been shown to affect various cellular members of the central nervous system in multitudinous ways, such as by modulating neuronal firing rates, inducing cellular apoptosis, or triggering synaptic pruning. These observations have in turn led to the exciting development of clinical therapies aiming to harness this neuro-immune interaction for the treatment of neuropsychiatric disease and symptoms. Besides the clinic, important theoretical fundamentals can be drawn from the immune system and applied to our understanding of the brain and neuropsychiatric disease. These new frameworks could lead to novel insights in the field and further potentiate the development of future therapies to treat neuropsychiatric disease.


Assuntos
Transtornos Neurocognitivos/imunologia , Neuroimunomodulação , Animais , Encéfalo , Citocinas , Humanos , Neurônios
6.
EMBO Mol Med ; 13(9): e14714, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34291577

RESUMO

Brain-matter vacuolation is a defining trait of all prion diseases, yet its cause is unknown. Here, we report that prion infection and prion-mimetic antibodies deplete the phosphoinositide kinase PIKfyve-which controls endolysosomal maturation-from mouse brains, cultured cells, organotypic brain slices, and brains of Creutzfeldt-Jakob disease victims. We found that PIKfyve is acylated by the acyltransferases zDHHC9 and zDHHC21, whose juxtavesicular topology is disturbed by prion infection, resulting in PIKfyve deacylation and rapid degradation, as well as endolysosomal hypertrophy and activation of TFEB-dependent lysosomal enzymes. A protracted unfolded protein response (UPR), typical of prion diseases, also induced PIKfyve deacylation and degradation. Conversely, UPR antagonists restored PIKfyve levels in prion-infected cells. Overexpression of zDHHC9 and zDHHC21, administration of the antiprion polythiophene LIN5044, or supplementation with the PIKfyve reaction product PI(3,5)P2 suppressed prion-induced vacuolation and restored lysosomal homeostasis. Thus, PIKfyve emerges as a central mediator of vacuolation and neurotoxicity in prion diseases.


Assuntos
Fosfatidilinositol 3-Quinases , Doenças Priônicas , Aciltransferases , Animais , Encéfalo/metabolismo , Homeostase , Lisossomos/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo
7.
Sci Transl Med ; 11(495)2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31167929

RESUMO

Parkinson's disease (PD) is a neurological disorder characterized by the progressive accumulation of neuronal α-synuclein (αSyn) inclusions called Lewy bodies. It is believed that Lewy bodies spread throughout the nervous system due to the cell-to-cell propagation of αSyn via cycles of secretion and uptake. Here, we investigated the internalization and intracellular accumulation of exogenous αSyn, two key steps of Lewy body pathogenesis, amplification and spreading. We found that stable αSyn fibrils substantially accumulate in different cell lines upon internalization, whereas αSyn monomers, oligomers, and dissociable fibrils do not. Our data indicate that the uptake-mediated accumulation of αSyn in a human-derived neuroblastoma cell line triggered an adaptive response that involved proteins linked to ubiquitin ligases of the S-phase kinase-associated protein 1 (SKP1), cullin-1 (Cul1), and F-box domain-containing protein (SCF) family. We found that SKP1, Cul1, and the F-box/LRR repeat protein 5 (FBXL5) colocalized and physically interacted with internalized αSyn in cultured cells. Moreover, the SCF containing the F-box protein FBXL5 (SCFFBXL5) catalyzed αSyn ubiquitination in reconstitution experiments in vitro using recombinant proteins and in cultured cells. In the human brain, SKP1 and Cul1 were recruited into Lewy bodies from brainstem and neocortex of patients with PD and related neurological disorders. In both transgenic and nontransgenic mice, intracerebral administration of exogenous αSyn fibrils triggered a Lewy body-like pathology, which was amplified by SKP1 or FBXL5 loss of function. Our data thus indicate that SCFFXBL5 regulates αSyn in vivo and that SCF ligases may constitute targets for the treatment of PD and other α-synucleinopathies.


Assuntos
Corpos de Lewy/metabolismo , Corpos de Lewy/patologia , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína/metabolismo , Animais , Benzotiazóis/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Camundongos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/metabolismo , Proteoma/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Ubiquitina/metabolismo
8.
PLoS One ; 12(2): e0170503, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28207746

RESUMO

Resistance to proteolytic digestion has long been considered a defining trait of prions in tissues of organisms suffering from transmissible spongiform encephalopathies. Detection of proteinase K-resistant prion protein (PrPSc) still represents the diagnostic gold standard for prion diseases in humans, sheep and cattle. However, it has become increasingly apparent that the accumulation of PrPSc does not always accompany prion infections: high titers of prion infectivity can be reached also in the absence of protease resistant PrPSc. Here, we describe a structural basis for the phenomenon of protease-sensitive prion infectivity. We studied the effect on proteinase K (PK) resistance of the amino acid substitution Y169F, which removes a single oxygen atom from the ß2-α2 loop of the cellular prion protein (PrPC). When infected with RML or the 263K strain of prions, transgenic mice lacking wild-type (wt) PrPC but expressing MoPrP169F generated prion infectivity at levels comparable to wt mice. The newly generated MoPrP169F prions were biologically indistinguishable from those recovered from prion-infected wt mice, and elicited similar pathologies in vivo. Surprisingly, MoPrP169F prions showed greatly reduced PK resistance and density gradient analyses showed a significant reduction in high-density aggregates. Passage of MoPrP169F prions into mice expressing wt MoPrP led to full recovery of protease resistance, indicating that no strain shift had taken place. We conclude that a subtle structural variation in the ß2-α2 loop of PrPC affects the sensitivity of PrPSc to protease but does not impact prion replication and infectivity. With these findings a specific structural feature of PrPC can be linked to a physicochemical property of the corresponding PrPSc.


Assuntos
Encéfalo/metabolismo , Endopeptidase K/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/transmissão , Príons/patogenicidade , Substituição de Aminoácidos , Animais , Encéfalo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Mutação/genética , Proteínas PrPSc/química , Proteínas PrPSc/genética , Doenças Priônicas/patologia , Conformação Proteica , Proteólise
9.
J Exp Med ; 213(6): 1047-59, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27185853

RESUMO

Microglial activation is a hallmark of most neurodegenerative disorders, and is particularly conspicuous in prion diseases. However, the role of microglia, which function as both primary immune effector cells and professional phagocytes in the central nervous system, remains contentious in the context of neurodegeneration. Here, we evaluated the effect of microglial depletion/deficiency on prion pathogenesis. We found that ganciclovir-mediated microglial ablation on tga20/CD11b-thymidine kinase of Herpes simplex virus (HSVTK) cerebellar organotypic cultured slices markedly aggravated prion-induced neurotoxicity. A similar deterioration of disease was recapitulated in in vivo microglial depletion in prion-infected tga20/CD11b-HSVTK mice. Additionally, deficiency of microglia in interleukin 34 knockout (IL34(-/-)) mice again resulted in significantly augmented proteinase K-resistant prion protein deposition and accelerated prion disease progression. These results provide unambiguous evidence for a general protective role of microglia in prion pathogenesis.


Assuntos
Interleucinas/metabolismo , Microglia/metabolismo , Doenças Priônicas/metabolismo , Príons/metabolismo , Animais , Interleucinas/genética , Camundongos , Camundongos Knockout , Microglia/patologia , Doenças Priônicas/genética , Doenças Priônicas/patologia , Príons/genética
10.
J Hepatol ; 65(1): 66-74, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26948495

RESUMO

BACKGROUND & AIMS: Liver can recover following resection. If tissue loss is too excessive, however, liver failure will develop as is known from the small-for-size-syndrome (SFSS). The molecular processes underlying liver failure are ill-understood. Here, we explored the role and the clinical potential of Nr1i3 (constitutive androstane receptor, Car) in liver failure following hepatectomy. METHODS: Activators of Car, various hepatectomies, Car(-/-) mice, humanized CAR mice, human tissue and ex vivo liver slice cultures were used to study Car in the SFSS. Pathways downstream of Car were investigated by in vivo siRNA knockdown. RESULTS: Excessive tissue loss causing liver failure is associated with deficient induction of Car. Reactivation of Car by an agonist normalizes all features associated with experimental SFSS. The beneficial effects of Car activation are relayed through Foxm1, an essential promoter of the hepatocyte cell cycle. Deficiency in the CAR-FOXM1 axis likewise is evident in human SFSS. Activation of human CAR mitigates SFSS in humanized CAR mice and improves the culture of human liver slices. CONCLUSIONS: Impaired hepatic Car-Foxm1 signaling provides a first molecular characterization of liver that fails to recover after tissue loss. Our findings place deficient regeneration as a principal cause behind the SFSS and suggest CAR agonists may bear clinical potential against liver failure. LAY SUMMARY: The unique regenerative capacity of liver has its natural limits. Following tissue loss that is too excessive, such as through extended resection in the clinic, liver failure may develop. This is known as small-for-size-syndrome (SFSS) and represents the most frequent cause of death due to liver surgery. Here we show that deficient induction of the protein Car, a central regulator of liver function and growth, is a cause of liver failure following extended resection; reactivation of Car through pharmacological means is sufficient to prevent or rescue the SFSS.


Assuntos
Falência Hepática , Animais , Receptor Constitutivo de Androstano , Hepatectomia , Humanos , Fígado , Regeneração Hepática , Camundongos , Receptores Citoplasmáticos e Nucleares
11.
PLoS Pathog ; 12(1): e1005401, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26821311

RESUMO

Antibodies against the prion protein PrPC can antagonize prion replication and neuroinvasion, and therefore hold promise as possible therapeutics against prion diseases. However, the safety profile of such antibodies is controversial. It was originally reported that the monoclonal antibody D13 exhibits strong target-related toxicity, yet a subsequent study contradicted these findings. We have reported that several antibodies against certain epitopes of PrPC, including antibody POM1, are profoundly neurotoxic, yet antibody ICSM18, with an epitope that overlaps with POM1, was reported to be innocuous when injected into mouse brains. In order to clarify this confusing situation, we assessed the neurotoxicity of antibodies D13 and ICSM18 with dose-escalation studies using diffusion-weighted magnetic resonance imaging and various histological techniques. We report that both D13 and ICSM18 induce rapid, dose-dependent, on-target neurotoxicity. We conclude that antibodies directed to this region may not be suitable as therapeutics. No such toxicity was found when antibodies against the flexible tail of PrPC were administered. Any attempt at immunotherapy or immunoprophylaxis of prion diseases should account for these potential untoward effects.


Assuntos
Anticorpos Monoclonais/toxicidade , Imunoterapia/métodos , Proteínas PrPC/imunologia , Doenças Priônicas/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epitopos de Linfócito B/imunologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Priônicas/patologia
12.
Sci Transl Med ; 7(299): 299ra123, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26246168

RESUMO

Prions cause transmissible spongiform encephalopathies for which no treatment exists. Prions consist of PrP(Sc), a misfolded and aggregated form of the cellular prion protein (PrP(C)). We explore the antiprion properties of luminescent conjugated polythiophenes (LCPs) that bind and stabilize ordered protein aggregates. By administering a library of structurally diverse LCPs to the brains of prion-infected mice via osmotic minipumps, we found that antiprion activity required a minimum of five thiophene rings bearing regularly spaced carboxyl side groups. Solid-state nuclear magnetic resonance analyses and molecular dynamics simulations revealed that anionic side chains interacted with complementary, regularly spaced cationic amyloid residues of model prions. These findings allowed us to extract structural rules governing the interaction between LCPs and protein aggregates, which we then used to design a new set of LCPs with optimized binding. The new set of LCPs showed robust prophylactic and therapeutic potency in prion-infected mice, with the lead compound extending survival by >80% and showing activity against both mouse and hamster prions as well as efficacy upon intraperitoneal administration into mice. These results demonstrate the feasibility of targeted chemical design of compounds that may be useful for treating diseases of aberrant protein aggregation such as prion disease.


Assuntos
Desenho de Fármacos , Polímeros , Doenças Priônicas/tratamento farmacológico , Tiofenos , Animais , Cricetinae , Espectroscopia de Ressonância Magnética , Camundongos , Simulação de Dinâmica Molecular , Polímeros/química , Polímeros/uso terapêutico , Relação Estrutura-Atividade , Tiofenos/química , Tiofenos/uso terapêutico
13.
PLoS Pathog ; 11(4): e1004808, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875479

RESUMO

[This corrects the article DOI: 10.1371/journal.ppat.1004662.].

14.
Neurobiol Aging ; 36(5): 1994-2003, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25816748

RESUMO

Dysfunctional variants of the innate immune cell surface receptor TREM2 (triggering receptor expressed on myeloid cells-2) were identified as major genetic risk factors for Alzheimer's disease and other neurodegenerative conditions. Here we assessed a possible involvement of TREM2 in prion disease. We report that TREM2 expression by microglia is significantly up-regulated upon prion infection. However, depletion of TREM2 did not affect disease incubation time and survival after intracerebral prion infection. Interestingly, markers of microglial activation were attenuated in prion-infected TREM2(-/-) mice, suggesting an involvement of TREM2 in prion-induced microglial activation. Further phenotype profiling of microglia revealed that TREM2 deficiency did not change microglial phenotypes. We conclude that TREM2 is involved in prion-induced microglial activation but does not noticeably modulate the pathogenesis of experimental prion infections.


Assuntos
Expressão Gênica/genética , Glicoproteínas de Membrana/fisiologia , Microglia/metabolismo , Microglia/patologia , Doenças Priônicas/genética , Receptores Imunológicos/fisiologia , Regulação para Cima/genética , Animais , Camundongos Transgênicos , Fenótipo , Fatores de Risco
15.
PLoS One ; 10(2): e0117412, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25658480

RESUMO

The cellular prion protein (PrPC) consists of a flexible N-terminal tail (FT, aa 23-128) hinged to a membrane-anchored globular domain (GD, aa 129-231). Ligation of the GD with antibodies induces rapid neurodegeneration, which is prevented by deletion or functional inactivation of the FT. Therefore, the FT is an allosteric effector of neurotoxicity. To explore its mechanism of action, we generated transgenic mice expressing the FT fused to a GPI anchor, but lacking the GD (PrPΔ141-225, or "FTgpi"). Here we report that FTgpi mice develop a progressive, inexorably lethal neurodegeneration morphologically and biochemically similar to that triggered by anti-GD antibodies. FTgpi was mostly retained in the endoplasmic reticulum, where it triggered a conspicuous unfolded protein response specifically activating the PERK pathway leading to phosphorylation of eIF2α and upregulation of CHOP ultimately leading to neurodegeration similar to what was observed in prion infection.


Assuntos
Cerebelo/patologia , Proteínas PrPC/metabolismo , Doenças Priônicas/metabolismo , Doenças Priônicas/patologia , Resposta a Proteínas não Dobradas , Animais , Cerebelo/metabolismo , Estresse do Retículo Endoplasmático , Camundongos , Camundongos Transgênicos , Proteínas PrPC/análise , Príons
16.
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
17.
Nature ; 501(7465): 102-6, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23903654

RESUMO

Prion infections cause lethal neurodegeneration. This process requires the cellular prion protein (PrP(C); ref. 1), which contains a globular domain hinged to a long amino-proximal flexible tail. Here we describe rapid neurotoxicity in mice and cerebellar organotypic cultured slices exposed to ligands targeting the α1 and α3 helices of the PrP(C) globular domain. Ligands included seven distinct monoclonal antibodies, monovalent Fab1 fragments and recombinant single-chain variable fragment miniantibodies. Similar to prion infections, the toxicity of globular domain ligands required neuronal PrP(C), was exacerbated by PrP(C) overexpression, was associated with calpain activation and was antagonized by calpain inhibitors. Neurodegeneration was accompanied by a burst of reactive oxygen species, and was suppressed by antioxidants. Furthermore, genetic ablation of the superoxide-producing enzyme NOX2 (also known as CYBB) protected mice from globular domain ligand toxicity. We also found that neurotoxicity was prevented by deletions of the octapeptide repeats within the flexible tail. These deletions did not appreciably compromise globular domain antibody binding, suggesting that the flexible tail is required to transmit toxic signals that originate from the globular domain and trigger oxidative stress and calpain activation. Supporting this view, various octapeptide ligands were not only innocuous to both cerebellar organotypic cultured slices and mice, but also prevented the toxicity of globular domain ligands while not interfering with their binding. We conclude that PrP(C) consists of two functionally distinct modules, with the globular domain and the flexible tail exerting regulatory and executive functions, respectively. Octapeptide ligands also prolonged the life of mice expressing the toxic PrP(C) mutant, PrP(Δ94-134), indicating that the flexible tail mediates toxicity in two distinct PrP(C)-related conditions. Flexible tail-mediated toxicity may conceivably play a role in further prion pathologies, such as familial Creutzfeldt-Jakob disease in humans bearing supernumerary octapeptides.


Assuntos
Anticorpos/imunologia , Anticorpos/toxicidade , Maleabilidade , Príons/química , Príons/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/toxicidade , Sítios de Ligação de Anticorpos , Calpaína/metabolismo , Cerebelo , Síndrome de Creutzfeldt-Jakob/metabolismo , Reagentes de Ligações Cruzadas , Mapeamento de Epitopos , Feminino , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/toxicidade , Técnicas In Vitro , Ligantes , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPC/imunologia , Príons/genética , Espécies Reativas de Oxigênio/metabolismo , Deleção de Sequência/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/toxicidade
18.
PLoS Pathog ; 8(11): e1002985, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133383

RESUMO

Prions cause neurodegeneration in vivo, yet prion-infected cultured cells do not show cytotoxicity. This has hampered mechanistic studies of prion-induced neurodegeneration. Here we report that prion-infected cultured organotypic cerebellar slices (COCS) experienced progressive spongiform neurodegeneration closely reproducing prion disease, with three different prion strains giving rise to three distinct patterns of prion protein deposition. Neurodegeneration did not occur when PrP was genetically removed from neurons, and a comprehensive pharmacological screen indicated that neurodegeneration was abrogated by compounds known to antagonize prion replication. Prion infection of COCS and mice led to enhanced fodrin cleavage, suggesting the involvement of calpains or caspases in pathogenesis. Accordingly, neurotoxicity and fodrin cleavage were prevented by calpain inhibitors but not by caspase inhibitors, whereas prion replication proceeded unimpeded. Hence calpain inhibition can uncouple prion replication from its neurotoxic sequelae. These data validate COCS as a powerful model system that faithfully reproduces most morphological hallmarks of prion infections. The exquisite accessibility of COCS to pharmacological manipulations was instrumental in recognizing the role of calpains in neurotoxicity, and significantly extends the collection of tools necessary for rigorously dissecting prion pathogenesis.


Assuntos
Cerebelo/metabolismo , Doenças Priônicas/metabolismo , Príons/metabolismo , Príons/patogenicidade , Animais , Calpaína/genética , Calpaína/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspases/genética , Caspases/metabolismo , Cerebelo/patologia , Camundongos , Camundongos Transgênicos , Microdissecção/métodos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Doenças Priônicas/genética , Doenças Priônicas/patologia , Príons/genética , Proteólise
19.
Clin Neurophysiol ; 122(10): 2025-31, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21474370

RESUMO

OBJECTIVES: To investigate whether there are any objective EEG characteristics that change significantly between specific time periods during maintenance of wakefulness test (MWT) and whether such changes are associated with the ability to appropriately communicate sleepiness. METHODS: After a night of total sleep deprivation, 12 healthy young subjects underwent a MWT whilst being instructed to communicate the experience of subjective sleepiness by pressing a button. EEG analysis consisted of average relative power and correlation between EEG signals. RESULTS: A comparison of the 30 s before microsleep (MS) with 30 s before subjects communicated experience of sleepiness (PB) showed increased ß correlation as well as increased power in the ß band (13-20 Hz) whereas power in the θ (4.5-7.5 Hz) and α (8-12.5 Hz) band was significantly decreased. When subjects later failed to communicate the experience of subjective sleepiness before (micro-)sleep occurred, average relative power and EEG correlation were significantly higher during 30 s following lights off in the δ (1-4 Hz) band and power in the α and ß bands was decreased. CONCLUSIONS: EEG spectral power and correlation change significantly in specific frequency bands between different time periods of MWT. Failure to communicate sleepiness is associated with certain precursors of EEG power and correlation. SIGNIFICANCE: This study demonstrates that there are specific EEG characteristics associated with impending failure to communicate sleepiness.


Assuntos
Eletroencefalografia/métodos , Privação do Sono/fisiopatologia , Sono/fisiologia , Vigília/fisiologia , Adulto , Feminino , Humanos , Masculino , Polissonografia/métodos , Adulto Jovem
20.
Sleep Med ; 11(8): 747-51, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20673742

RESUMO

OBJECTIVE: To test whether subjects spontaneously signal sleepiness before falling asleep under monotonous conditions. METHODS: Twenty-eight healthy students were deprived of sleep for one night and then underwent a "maintenance-of-wakefulness test" (MWT) consisting of four 40-min trials. They were told to give a signal as soon as they felt sleepy and to try to stay awake as long as possible. In a first series of tests, the subjects were given no reward (nr); in a second series, monetary rewards (wr) were given both for an accurate perception of sleepiness and for staying awake longer. RESULTS: Seventeen of the 28 subjects (60.7%) did not signal sleepiness before a sleep fragment occurred in at least one of the four MWT trials. Women were more reliably aware of sleepiness than men in the nr trials (p=.02), while the men's performance improved in the wr trials (p<.02), becoming equivalent to the women's performance. CONCLUSIONS: Our results cast doubt on the general assumption that one cannot fall asleep without feeling sleepy first. If similar results can be obtained in monotonous driving or working situations, this will imply that accidents caused by sleepiness or by falling asleep cannot necessarily be attributed to an individual's negligence.


Assuntos
Conscientização/fisiologia , Percepção/fisiologia , Privação do Sono/fisiopatologia , Fases do Sono/fisiologia , Acidentes , Acidentes de Trânsito , Adulto , Feminino , Humanos , Masculino , Motivação , Recompensa , Fatores de Risco , Distribuição por Sexo , Privação do Sono/epidemiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...