Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Biomater Sci Eng ; 10(3): 1856-1868, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38385618

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disease and the most frequent cause of dementia. It is characterized by the accumulation in the brain of two pathological protein aggregates: amyloid-ß peptides (Aß) and abnormally phosphorylated tau. The progressive cognitive decline observed in patients strongly correlates with the synaptic loss. Many lines of evidence suggest that soluble forms of Aß accumulate into the brain where they cause synapse degeneration. Stopping their spreading and/or targeting the pathophysiological mechanisms leading to synaptic loss would logically be beneficial for the patients. However, we are still far from understanding these processes. Our objective was therefore to develop a versatile model to assay and study Aß-induced synaptotoxicity. We integrated a microfluidic device that physically isolates synapses from presynaptic and postsynaptic neurons with a microelectrode array. We seeded mouse primary cortical cells in the presynaptic and postsynaptic chambers. After functional synapses have formed in the synaptic chamber, we exposed them to concentrated conditioned media from cell lines overexpressing the wild-type or mutated amyloid precursor protein and thus secreting different levels of Aß. We recorded the neuronal activity before and after exposition to Aß and quantified Aß's effects on the connectivity between presynaptic and postsynaptic neurons. We observed that the application of Aß on the synapses for 48 h strongly decreased the interchamber connectivity without significantly affecting the neuronal activity in the presynaptic or postsynaptic chambers. Thus, through this model, we are able to functionally assay the impact of Aß peptides (or other molecules) on synaptic connectivity and to use the latter as a proxy to study Aß-induced synaptotoxicity. Moreover, since the presynaptic, postsynaptic, and synaptic chambers can be individually targeted, our assay provides a powerful tool to evaluate the involvement of candidate genes in synaptic vulnerability and/or test therapeutic strategies for AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Ratones , Animales , Humanos , Microelectrodos , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Dispositivos Laboratorio en un Chip
2.
Biomaterials ; 305: 122426, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38134473

RESUMEN

Microglial cells, as the primary defense line in the central nervous system, play a crucial role in responding to various mechanical signals that can trigger their activation. Despite extensive research on the impact of chemical signaling on brain cells, the understanding of mechanical signaling in microglia remains limited. To bridge this gap, we subjected microglial cells to a singular mechanical stretch and compared their responses with those induced by lipopolysaccharide treatment, a well-established chemical activator. Here we show that stretching microglial cells leads to their activation, highlighting their significant mechanosensitivity. Stretched microglial cells exhibited distinct features, including elevated levels of Iba1 protein, a denser actin cytoskeleton, and increased persistence in migration. Unlike LPS-treated microglial cells, the secretory profile of chemokines and cytokines remained largely unchanged in response to stretching, except for TNF-α. Intriguingly, a single stretch injury resulted in more compacted chromatin and DNA damage, suggesting potential long-term genomic instabilities in stretched microglia. Using compartmentalized microfluidic chambers with neuronal networks, we observed that stretched microglial cells exhibited enhanced phagocytic and synaptic stripping activities. These findings collectively suggest that stretching events can unlock the immune potential of microglial cells, contributing to the maintenance of brain tissue homeostasis following mechanical injury.


Asunto(s)
Microglía , Fagocitos , Microglía/metabolismo , Sistema Nervioso Central , Encéfalo , Transducción de Señal , Lipopolisacáridos/farmacología
3.
Biosens Bioelectron ; 237: 115538, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37506488

RESUMEN

Microelectrode Arrays (MEAs) are popular tools for in vitro extracellular recording. They are often optimized by surface engineering to improve affinity with neurons and guarantee higher recording quality and stability. Recently, PEDOT:PSS has been used to coat microelectrodes due to its good biocompatibility and low impedance, which enhances neural coupling. Herein, we investigate on electro-co-polymerization of EDOT with its triglymated derivative to control valence between monomer units and hydrophilic functions on a conducting polymer. Molecular packing, cation complexation, dopant stoichiometry are governed by the glycolation degree of the electro-active coating of the microelectrodes. Optimal monomer ratio allows fine-tuning the material hydrophilicity and biocompatibility without compromising the electrochemical impedance of microelectrodes nor their stability while interfaced with a neural cell culture. After incubation, sensing readout on the modified electrodes shows higher performances with respect to unmodified electropolymerized PEDOT, with higher signal-to-noise ratio (SNR) and higher spike counts on the same neural culture. Reported SNR values are superior to that of state-of-the-art PEDOT microelectrodes and close to that of state-of-the-art 3D microelectrodes, with a reduced fabrication complexity. Thanks to this versatile technique and its impact on the surface chemistry of the microelectrode, we show that electro-co-polymerization trades with many-compound properties to easily gather them into single macromolecular structures. Applied on sensor arrays, it holds great potential for the customization of neurosensors to adapt to environmental boundaries and to optimize extracted sensing features.


Asunto(s)
Técnicas Biosensibles , Microelectrodos , Electrodos Implantados , Polímeros/química , Neuronas/fisiología
4.
Biomed Phys Eng Express ; 9(3)2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36745905

RESUMEN

Recently, the development of electronic devices to extracellularly record the simultaneous electrical activities of numerous neurons has been blooming, opening new possibilities to interface and decode neuronal activity. In this work, we tested how the use of EDOT electropolymerization to tune post-fabrication materials could optimize the cell/electrode interface of such devices. Our results showed an improved signal-to-noise ratio, better biocompatibility, and a higher number of neurons detected in comparison with gold electrodes. Then, using such enhanced recordings with 2D neuronal cultures combined with fluorescent optical imaging, we checked the extent to which the positions of the recorded neurons could be estimated solely via their extracellular signatures. Our results showed that assuming neurons behave as monopoles, positions could be estimated with a precision of approximately tens of micrometers.


Asunto(s)
Técnicas de Cultivo de Célula , Neuronas , Microelectrodos , Potenciales de Acción/fisiología , Neuronas/fisiología , Oro
5.
J Neural Eng ; 19(6)2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36374001

RESUMEN

Objective.Tau ablation has a protective effect in epilepsy due to inhibition of the hyperexcitability/hypersynchrony. Protection may also occur in transgenic models of Alzheimer's disease by reducing the epileptic activity and normalizing the excitation/inhibition imbalance. However, it is difficult to determine the exact functions of tau, because tau knockout (tauKO) brain networks exhibit elusive phenotypes. In this study, we aimed to further explore the physiological role of tau using brain network remodeling.Approach.The effect of tau ablation was investigated in hippocampal-entorhinal slice co-cultures during network remodeling. We recorded the spontaneous extracellular neuronal activity over 2 weeks in single-slice cultures and co-cultures from control andtauKOmice. We compared the burst activity and applied concepts and analytical tools intended for the analysis of the network synchrony and connectivity.Main results.Comparison of the control andtauKOco-cultures revealed that tau ablation had an anti-synchrony effect on the hippocampal-entorhinal two-slice networks at late stages of culture, in line with the literature. Differences were also found between the single-slice and co-culture conditions, which indicated that tau ablation had differential effects at the sub-network scale. For instance, tau ablation was found to have an anti-synchrony effect on the co-cultured hippocampal slices throughout the culture, possibly due to a reduction in the excitation/inhibition ratio. Conversely, tau ablation led to increased synchrony in the entorhinal slices at early stages of the co-culture, possibly due to homogenization of the connectivity distribution.Significance.The new methodology presented here proved useful for investigating the role of tau in the remodeling of complex brain-derived neural networks. The results confirm previous findings and hypotheses concerning the effects of tau ablation on neural networks. Moreover, the results suggest, for the first time, that tau has multifaceted roles that vary in different brain sub-networks.


Asunto(s)
Epilepsia , Neuronas , Animales , Ratones , Técnicas de Cocultivo , Encéfalo , Hipocampo , Redes Neurales de la Computación
6.
ACS Chem Neurosci ; 12(20): 3885-3897, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34614352

RESUMEN

Deformation, compression, or stretching of brain tissues cause diffuse axonal injury (DAI) and induce structural and functional alterations of astrocytes, the most abundant cell type in the brain. To gain further insight into the role of mechanically activated astrocytes on neuronal networks, this study was designed to investigate whether cytokines released by mechanically activated astrocytes can affect the growth and synaptic connections of cortical neuronal networks. Astrocytes were cultivated on elastic membranes and subjected to repetitive mechanical insults, whereas well-defined protein micropatterns were used to form standardized neuronal networks. GFAP staining showed that astrocytes were mechanically activated after two cycles of stretch and mesoscale discovery assays indicated that injured astrocytes released four major cytokines. To understand the role of these cytokines, neuronal networks were cultured with the supernatant of healthy or mechanically activated astrocytes, and the individual contribution of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) was studied. We found that the supernatant of two-cycle stretched astrocytes decreased presynaptic terminals and indicated that TNF-α must be considered a key player of the synaptic loss. Furthermore, our results indicate that cytokines released by injured astrocytes significantly modulate the balance between TNFR1 and TNFR2 receptors by enhancing R2 receptors. We demonstrated that TNF-α is not involved in this process, suggesting a predominant role of other secreted cytokines. Together, these results contribute to a better understanding of the consequences of repetitive astrocyte deformations and highlight the role of inflammatory signaling pathways in synaptic plasticity and modulation of TNFR1 and TNFR2 receptors.


Asunto(s)
Astrocitos , Receptores Tipo II del Factor de Necrosis Tumoral , Células Cultivadas , Citocinas , Humanos , Factor de Necrosis Tumoral alfa
7.
Neurotherapeutics ; 18(2): 1137-1150, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33533011

RESUMEN

Prion diseases are caused by the propagation of PrPSc, the pathological conformation of the PrPC prion protein. The molecular mechanisms underlying PrPSc propagation are still unsolved and no therapeutic solution is currently available. We thus sought to identify new anti-prion molecules and found that flunarizine inhibited PrPSc propagation in cell culture and significantly prolonged survival of prion-infected mice. Using an in silico therapeutic repositioning approach based on similarities with flunarizine chemical structure, we tested azelastine, duloxetine, ebastine, loperamide and metixene and showed that they all have an anti-prion activity. Like flunarizine, these marketed drugs reduced PrPSc propagation in cell culture and in mouse cerebellum organotypic slice culture, and inhibited the protein folding activity of the ribosome (PFAR). Strikingly, some of these drugs were also able to alleviate phenotypes due to PABPN1 nuclear aggregation in cell and Drosophila models of oculopharyngeal muscular dystrophy (OPMD). These data emphasize the therapeutic potential of anti-PFAR drugs for neurodegenerative and neuromuscular proteinopathies.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Flunarizina/administración & dosificación , Proteína I de Unión a Poli(A)/metabolismo , Enfermedades por Prión/metabolismo , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Línea Celular , Bases de Datos Factuales , Drosophila , Femenino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Proteína I de Unión a Poli(A)/antagonistas & inhibidores , Proteína I de Unión a Poli(A)/genética , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/genética , Proteínas Priónicas/antagonistas & inhibidores , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Agregado de Proteínas/fisiología , Ovinos
8.
Nat Neurosci ; 23(12): 1567-1579, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33169029

RESUMEN

Alzheimer's disease (AD) is characterized by the accumulation of the tau protein in neurons, neurodegeneration and memory loss. However, the role of non-neuronal cells in this chain of events remains unclear. In the present study, we found accumulation of tau in hilar astrocytes of the dentate gyrus of individuals with AD. In mice, the overexpression of 3R tau specifically in hilar astrocytes of the dentate gyrus altered mitochondrial dynamics and function. In turn, these changes led to a reduction of adult neurogenesis, parvalbumin-expressing neurons, inhibitory synapses and hilar gamma oscillations, which were accompanied by impaired spatial memory performances. Together, these results indicate that the loss of tau homeostasis in hilar astrocytes of the dentate gyrus is sufficient to induce AD-like symptoms, through the impairment of the neuronal network. These results are important for our understanding of disease mechanisms and underline the crucial role of astrocytes in hippocampal function.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Astrocitos/metabolismo , Giro Dentado/metabolismo , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/psicología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/complicaciones , Animales , Animales Modificados Genéticamente , Femenino , Humanos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/metabolismo , Neurogénesis , Parvalbúminas/metabolismo , Embarazo , Desempeño Psicomotor , Ratas , Memoria Espacial , Sinapsis/fisiología
9.
Mol Neurobiol ; 56(3): 2159-2173, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29998397

RESUMEN

Among the ever-growing number of self-replicating proteins involved in neurodegenerative diseases, the prion protein PrP remains the most infamous for its central role in transmissible spongiform encephalopathies (TSEs). In these diseases, pathogenic prions propagate through a seeding mechanism, where normal PrPC molecules are converted into abnormally folded scrapie isoforms termed PrPSc. Since its discovery over 30 years ago, much advance has contributed to define the host-encoded cellular prion protein PrPC as a critical relay of prion-induced neuronal cell demise. A current consensual view is that the conversion of PrPC into PrPSc in neuronal cells diverts the former from its normal function with subsequent molecular alterations affecting synaptic plasticity. Here, we report that prion infection is associated with reduced expression of key effectors of the Notch pathway in vitro and in vivo, recapitulating changes fostered by the absence of PrPC. We further show that both prion infection and PrPC depletion promote drastic alterations in the expression of a defined set of Eph receptors and their ephrin ligands, which represent important players in synaptic function. Our data indicate that defects in the Notch and Eph axes can be mitigated in response to histone deacetylase inhibition in PrPC-depleted as well as prion-infected cells. We thus conclude that infectious prions cause a loss-of-function phenotype with respect to Notch and Eph signaling and that these alterations are sustained by epigenetic mechanisms.


Asunto(s)
Enfermedades por Prión/metabolismo , Proteínas Priónicas/metabolismo , Receptores de la Familia Eph/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Epigénesis Genética , Ratones , Neuronas/metabolismo , Enfermedades por Prión/genética
10.
Stem Cells ; 35(3): 754-765, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27641601

RESUMEN

The prion protein is infamous for its involvement in a group of neurodegenerative diseases known as Transmissible Spongiform Encephalopathies. In the longstanding quest to decipher the physiological function of its cellular isoform, PrPC , the discovery of its participation to the self-renewal of hematopoietic and neural stem cells has cast a new spotlight on its potential role in stem cell biology. However, still little is known on the cellular and molecular mechanisms at play. Here, by combining in vitro and in vivo murine models of PrPC depletion, we establish that PrPC deficiency severely affects the Notch pathway, which plays a major role in neural stem cell maintenance. We document that the absence of PrPC in a neuroepithelial cell line or in primary neurospheres is associated with drastically reduced expression of Notch ligands and receptors, resulting in decreased levels of Notch target genes. Similar alterations of the Notch pathway are recovered in the neuroepithelium of Prnp-/- embryos during a developmental window encompassing neural tube closure. In addition, in line with Notch defects, our data show that the absence of PrPC results in altered expression of Nestin and Olig2 as well as N-cadherin distribution. We further provide evidence that PrPC controls the expression of the epidermal growth factor receptor (EGFR) downstream from Notch. Finally, we unveil a negative feedback action of EGFR on both Notch and PrPC . As a whole, our study delineates a molecular scenario through which PrPC takes part to the self-renewal of neural stem and progenitor cells. Stem Cells 2017;35:754-765.


Asunto(s)
Células-Madre Neurales/metabolismo , Proteínas Priónicas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Biomarcadores/metabolismo , Cadherinas/metabolismo , Comunicación Celular , Línea Celular , Linaje de la Célula , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Receptores ErbB/metabolismo , Retroalimentación Fisiológica , Ratones
11.
Sci Rep ; 5: 17146, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26679898

RESUMEN

Although conversion of the cellular form of the prion protein (PrP(C)) into a misfolded isoform is the underlying cause of prion diseases, understanding PrP(C) physiological functions has remained challenging. PrP(C) depletion or overexpression alters the proliferation and differentiation properties of various types of stem and progenitor cells in vitro by unknown mechanisms. Such involvement remains uncertain in vivo in the absence of any drastic phenotype of mice lacking PrP(C). Here, we report PrP(C) enrichment at the base of the primary cilium in stem and progenitor cells from the central nervous system and cardiovascular system of developing mouse embryos. PrP(C) depletion in a neuroepithelial cell line dramatically altered key cilium-dependent processes, such as Sonic hedgehog signalling and α-tubulin post-translational modifications. These processes were also affected over a limited time window in PrP(C)-ablated embryos. Thus, our study reveals PrP(C) as a potential actor in the developmental regulation of microtubule dynamics and ciliary functions.


Asunto(s)
Cilios/metabolismo , Priones/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Células Cultivadas , Sistema Nervioso Central/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Proteínas Hedgehog/metabolismo , Ratones , Microscopía Confocal , Proteínas PrPC/deficiencia , Proteínas PrPC/genética , Priones/genética , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
12.
Nat Commun ; 6: 6894, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25902731

RESUMEN

Caprine-like Generalized Hypoplasia Syndrome (SHGC) is an autosomal-recessive disorder in Montbéliarde cattle. Affected animals present a wide range of clinical features that include the following: delayed development with low birth weight, hind limb muscular hypoplasia, caprine-like thin head and partial coat depigmentation. Here we show that SHGC is caused by a truncating mutation in the CEP250 gene that encodes the centrosomal protein C-Nap1. This mutation results in centrosome splitting, which neither affects centriole ultrastructure and duplication in dividing cells nor centriole function in cilium assembly and mitotic spindle organization. Loss of C-Nap1-mediated centriole cohesion leads to an altered cell migration phenotype. This discovery extends the range of loci that constitute the spectrum of autosomal primary recessive microcephaly (MCPH) and Seckel-like syndromes.


Asunto(s)
Enfermedades de los Bovinos/genética , Proteínas de Ciclo Celular/genética , Movimiento Celular/genética , Centriolos/metabolismo , Hipopigmentación/veterinaria , Microcefalia/veterinaria , Morfogénesis/genética , Enfermedades Musculares/veterinaria , Animales , Bovinos , Hipopigmentación/genética , Microcefalia/genética , Enfermedades Musculares/genética , Mutación , Síndrome
13.
Front Cell Dev Biol ; 2: 35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25364742

RESUMEN

Based on its developmental pattern of expression, early studies suggested the implication of the mammalian Prion protein PrP, a glycosylphosphatidylinositol-anchored ubiquitously expressed and evolutionary conserved glycoprotein encoded by the Prnp gene, in early embryogenesis. However, gene invalidation in several species did not result in obvious developmental abnormalities and it was only recently that it was associated in mice with intra-uterine growth retardation and placental dysfunction. A proposed explanation for this lack of easily detectable developmental-related phenotype is the existence in the genome of one or more gene (s) able to compensate for the absence of PrP. Indeed, two other members of the Prnp gene family have been recently described, Doppel and Shadoo, and the consequences of their invalidation alongside that of PrP tested in mice. No embryonic defect was observed in mice depleted for Doppel and PrP. Interestingly, the co-invalidation of PrP and Shadoo in two independent studies led to apparently conflicting observations, with no apparent consequences in one report and the observation of a developmental defect of the ectoplacental cone that leads to early embryonic lethality in the other. This short review aims at summarizing these recent, apparently conflicting data highlighting the related biological questions and associated implications in terms of animal and human health.

14.
Front Cell Dev Biol ; 2: 55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25364760

RESUMEN

The cellular prion protein PrP(C) was initially discovered as the normal counterpart of the pathological scrapie prion protein PrP(Sc), the main component of the infectious agent of Transmissible Spongiform Encephalopathies. While clues as to the physiological function of this ubiquitous protein were greatly anticipated from the development of knockout animals, PrP-null mice turned out to be viable and to develop without major phenotypic abnormalities. Notwithstanding, the discovery that hematopoietic stem cells from PrP-null mice have impaired long-term repopulating potential has set the stage for investigating into the role of PrP(C) in stem cell biology. A wealth of data have now exemplified that PrP(C) is expressed in distinct types of stem cells and regulates their self-renewal as well as their differentiation potential. A role for PrP(C) in the fate restriction of embryonic stem cells has further been proposed. Paralleling these observations, an overexpression of PrP(C) has been documented in various types of tumors. In line with the contribution of PrP(C) to stemness and to the proliferation of cancer cells, PrP(C) was recently found to be enriched in subpopulations of tumor-initiating cells. In the present review, we summarize the current knowledge of the role played by PrP(C) in stem cell biology and discuss how the subversion of its function may contribute to cancer progression.

15.
Front Cell Dev Biol ; 2: 58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25364763

RESUMEN

The deletion of the cellular form of the prion protein (PrP(C)) in mouse, goat, and cattle has no drastic phenotypic consequence. This stands in apparent contradiction with PrP(C) quasi-ubiquitous expression and conserved primary and tertiary structures in mammals, and its pivotal role in neurodegenerative diseases such as prion and Alzheimer's diseases. In zebrafish embryos, depletion of PrP ortholog leads to a severe loss-of-function phenotype. This raises the question of a potential role of PrP(C) in the development of all vertebrates. This view is further supported by the early expression of the PrP(C) encoding gene (Prnp) in many tissues of the mouse embryo, the transient disruption of a broad number of cellular pathways in early Prnp(-/-) mouse embryos, and a growing body of evidence for PrP(C) involvement in the regulation of cell proliferation and differentiation in various types of mammalian stem cells and progenitors. Finally, several studies in both zebrafish embryos and in mammalian cells and tissues in formation support a role for PrP(C) in cell adhesion, extra-cellular matrix interactions and cytoskeleton. In this review, we summarize and compare the different models used to decipher PrP(C) functions at early developmental stages during embryo- and organo-genesis and discuss their relevance.

16.
ACS Chem Neurosci ; 5(10): 1075-82, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25244284

RESUMEN

Guanabenz (GA) is an orally active α2-adrenergic agonist that has been used for many years for the treatment of hypertension. We recently described that GA is also active against both yeast and mammalian prions in an α2-adrenergic receptor-independent manner. These data suggest that this side-activity of GA could be explored for the treatment of prion-based diseases and other amyloid-based disorders. In this perspective, the potent antihypertensive activity of GA happens to be an annoying side-effect that could limit its use. In order to get rid of GA agonist activity at α2-adrenergic receptors, we performed a structure-activity relationship study around GA based on changes of the chlorine positions on the benzene moiety and then on the modifications of the guanidine group. Hence, we identified the two derivatives 6 and 7 that still possess a potent antiprion activity but were totally devoid of any agonist activity at α2-adrenergic receptors. Similarly to GA, 6 and 7 were also able to inhibit the protein folding activity of the ribosome (PFAR) which has been suggested to be involved in prion appearance/maintenance. Therefore, these two GA derivatives are worth being considered as drug candidates.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Guanabenzo/análogos & derivados , Guanabenzo/farmacología , Fármacos Neuroprotectores/farmacología , Priones/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 2/química , Animales , Células CHO , Bovinos , Cerebelo/efectos de los fármacos , Cerebelo/fisiopatología , Cricetulus , Escherichia coli , Guanabenzo/química , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Estructura Molecular , Fármacos Neuroprotectores/química , Proteínas PrPSc/metabolismo , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/fisiopatología , Pliegue de Proteína/efectos de los fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo , Relación Estructura-Actividad , Técnicas de Cultivo de Tejidos , Levaduras
17.
PLoS One ; 9(8): e104287, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25122456

RESUMEN

Prion transmission can occur by blood transfusion in human variant Creutzfeldt-Jakob disease and in experimental animal models, including sheep. Screening of blood and its derivatives for the presence of prions became therefore a major public health issue. As infectious titer in blood is reportedly low, highly sensitive and robust methods are required to detect prions in blood and blood derived products. The objectives of this study were to compare different methods--in vitro, ex vivo and in vivo assays--to detect prion infectivity in cells prepared from blood samples obtained from scrapie infected sheep at different time points of the disease. Protein misfolding cyclic amplification (PMCA) and bioassays in transgenic mice expressing the ovine prion protein were the most efficient methods to identify infected animals at any time of the disease (asymptomatic to terminally-ill stages). However scrapie cell and cerebellar organotypic slice culture assays designed to replicate ovine prions in culture also allowed detection of prion infectivity in blood cells from asymptomatic sheep. These findings confirm that white blood cells are appropriate targets for preclinical detection and introduce ex vivo tools to detect blood infectivity during the asymptomatic stage of the disease.


Asunto(s)
Bioensayo/métodos , Leucocitos/química , Priones/patogenicidad , Scrapie/diagnóstico , Animales , Infecciones Asintomáticas , Cerebelo/metabolismo , Ratones , Ratones Transgénicos , Proteínas PrPSc/metabolismo , Scrapie/metabolismo , Ovinos/metabolismo
18.
J Virol ; 88(15): 8678-86, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850746

RESUMEN

UNLABELLED: The dietary exposure of the human population to the prions responsible for the bovine spongiform encephalopathy (BSE) epizooty has led to the emergence of variant Creutzfeldt-Jakob disease (vCJD). This fatal, untreatable neurodegenerative disorder is a growing public health concern because the prevalence of the infection seems much greater than the disease incidence and because secondary transmission of vCJD by blood transfusion or use of blood products has occurred. A current limitation in variant CJD risk assessment is the lack of quantitative information on the infectivity of contaminated tissues. To address this limitation, we tested the potential of a transgenic mouse line overexpressing human prion protein (PrP), which was previously reported to propagate vCJD prions. Endpoint titration of vCJD infectivity in different tissues was evaluated by two different methods: (i) the "classical" bioassay, based on the appearance of clinical symptoms and the detection of pathological prion protein in tissues of the inoculated mouse, and (ii) a shortened bioassay based on the detection of the protein in the mouse spleen at defined time points. The two methods proved equally sensitive in quantifying infectivity, even after very-low-dose inoculation of infected material, but the time schedule was shortened from ~2.5 years to ~1 year with the spleen bioassay. Compared to the "gold-standard" RIII model routinely used for endpoint titration of vCJD/BSE prions, either method improved the sensitivity by >2 orders of magnitude and allowed reevaluating the infectious titer of spleen from a vCJD individual at disease end stage to >1,000-fold-higher values. IMPORTANCE: Here, we provide key reevaluation of the infectious titer of variant CJD brain and spleen tissues. The highly sensitive, accelerated spleen-based assay should thus constitute a key advance for variant CJD epidemiological and risk assessment purposes and should greatly facilitate future titration studies, including, for example, those aimed at validating decontamination procedures. The overlooked notion that the lymphoid tissue exhibits a higher capacity than the brain to replicate prions even after low-dose infection raises new questions about the molecular and/or cellular determinant(s) involved, a key issue regarding potent silent carriers of variant CJD in the lymphoid tissue.


Asunto(s)
Técnicas de Laboratorio Clínico/métodos , Síndrome de Creutzfeldt-Jakob/diagnóstico , Priones/análisis , Bazo/química , Animales , Bioensayo , Humanos , Ratones , Ratones Transgénicos , Sensibilidad y Especificidad , Factores de Tiempo
19.
J Gen Virol ; 94(Pt 6): 1435-1440, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23388201

RESUMEN

In naturally acquired transmissible spongiform encephalopathies, the pathogenic agents or prions spread from the sites of initial peripheral uptake or replication to the brain where they cause progressive and fatal neurodegeneration. Routing via the peripheral nervous system is considered to be one of the main pathways to the central nervous system. Replication of prions in Schwann cells is viewed as a potentially important mechanism for efficient prion spread along nerves. Here we used a Cre-loxP mouse transgenetic approach to disrupt host-encoded prion protein (PrP(C)) specifically in myelinating Schwann cells. Despite the use of infection routes targeting highly myelinated nerves, there was no alteration in mouse prion pathogenesis, suggesting that conversion-dependent, centripetal spread of prions does not crucially rely on PrP(C) expressed by myelinating Schwann cells.


Asunto(s)
Marcación de Gen , Vaina de Mielina/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Células de Schwann/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/patología
20.
Prion ; 7(2): 127-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23154632

RESUMEN

Although the pivotal implication of the host-encoded Prion protein, PrP, in the neuropathology of transmissible spongiform encephalopathy is known for decades, its biological role remains mostly elusive. Genetic inactivation is one way to assess such issue but, so far, PrP-knockout mice did not help much. However, recent reports involving (1) further studies of these mice during embryogenesis, (2) knockdown experiments in Zebrafish and (3) knockdown of Shadoo, a protein with PrP-like functional domains, in PrP-knockout mice, all suggested a role of the Prion protein family in early embryogenesis. This view is challenged by the recent report that PrP/Shadoo knockout mice are healthy and fertile. Although puzzling, these apparently contradictory data may on the contrary help at deciphering the Prion protein family role through focusing scientific attention outside the central nervous system and by helping the identification of other loci involved in the genetic robustness associated with PrP.


Asunto(s)
Priones/metabolismo , Animales , Desarrollo Embrionario , Humanos , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...