Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell Mol Neurobiol ; 42(5): 1429-1440, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33462779

RESUMEN

Rabies virus (RABV) is a neurotropic virus exclusively infecting neurons in the central nervous system. RABV encodes five proteins. Among them, the viral glycoprotein (RVG) plays a key role in viral entry into neurons and rabies pathogenesis. It was shown that the nature of the C-terminus of the RABV G protein, which possesses a PDZ-binding motif (PBM), modulates the virulence of the RABV strain. The neuronal protein partners recruited by this PBM may alter host cell function. This study was conducted to investigate the effect of RVG on synaptic function in the hippocampal dentate gyrus (DG) of rat. Two µl (108 T.U./ml) of the lentiviral vector containing RVG gene was injected into the DG of rat hippocampus. After 2 weeks, the rat's brain was cross-sectioned and RVG-expressing cells were detected by fluorescent microscopy. Hippocampal synaptic activity of the infected rats was then examined by recording the local field potentials from DG after stimulation of the perforant pathway. Short-term synaptic plasticity was also assessed by double pulse stimulation. Expression of RVG in DG increased long-term potentiation population spikes (LTP-PS), whereas no facilitation of LTP-PS was found in neurons expressing δRVG (deleted PBM). Furthermore, RVG and δRVG strengthened paired-pulse facilitation. Heterosynaptic long-term depression (LTD) in the DG was significantly blocked in RVG-expressing group compared to the control group. This blockade was dependent to PBM motif as rats expressing δRVG in the DG-expressed LTD comparable to the RVG group. Our data demonstrate that RVG expression facilitates both short- and long-term synaptic plasticity in the DG indicating that it may involve both pre- and postsynaptic mechanisms to alter synaptic function. Further studies are needed to elucidate the underlying mechanisms.


Asunto(s)
Virus de la Rabia , Animales , Giro Dentado/metabolismo , Estimulación Eléctrica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicoproteínas/farmacología , Hipocampo/metabolismo , Potenciación a Largo Plazo , Plasticidad Neuronal/fisiología , Virus de la Rabia/metabolismo , Ratas
2.
J Neurovirol ; 27(3): 434-443, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33788140

RESUMEN

Rabies is a life-threatening viral infection of the brain. Rabies virus (RABV) merely infects excitable cells including neurons provoking drastic behaviors including negative emotional memories. RABV glycoprotein (RVG) plays a critical role in RABV pathogenesis. RVG interacts with various cytoplasmic PDZ (PSD-95/Dlg/ZO-1) containing proteins through its PDZ binding motif (PBM). PTZ domains have crucial role in formation and function of signal transduction. Hippocampus is one of the cerebral regions that contain high load of viral antigens. We examined impact of RVG expression in the dorsal hippocampus on aversive as well as spatial learning and memory performance in rats. Two microliter of the lentiviral vector (~108 T.U./ml) encoding RVG or ∆RVG (deleted PBM) genomes was microinjected into the hippocampal CA1. After 1 week, rat's brain was cross-sectioned and RVG/∆RVG-expressing neuronal cells were confirmed by fluorescent microscopy. Passive avoidance and spatial learning and memory were assessed in rats by Shuttle box and Morris water maze (MWM). In the shuttle box, both RVG and ∆RVG decreased the time spent in the dark compartment compared to control (p < 0.05). In MWM, RVG and ∆RVG did not affect the acquisition of spatial task. In the probe test, RVG-expressing rats spent more time in the target quadrant, and also reached the platform position sooner than control group (p < 0.05). Rats expressing ∆RVG significantly swam farther from the hidden platform than RVG group (p < 0.05). Our data indicate RVG expression in the hippocampus strengthens aversive and spatial learning and memory performance. The boosting effect on spatial but not avoidance memory is mediated through PBM.


Asunto(s)
Reacción de Prevención , Región CA1 Hipocampal/fisiopatología , Glicoproteínas/genética , Aprendizaje por Laberinto , Virus de la Rabia/genética , Memoria Espacial , Proteínas Virales/genética , Animales , Región CA1 Hipocampal/metabolismo , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glicoproteínas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inyecciones Intraventriculares , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Virus de la Rabia/química , Virus de la Rabia/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Técnicas Estereotáxicas , Transgenes , Proteínas Virales/metabolismo
3.
J Biol Chem ; 294(37): 13755-13768, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346033

RESUMEN

Protection of neuronal homeostasis is a major goal in the management of neurodegenerative diseases. Microtubule-associated Ser/Thr kinase 2 (MAST2) inhibits neurite outgrowth, and its inhibition therefore represents a potential therapeutic strategy. We previously reported that a viral protein (G-protein from rabies virus) capable of interfering with protein-protein interactions between the PDZ domain of MAST2 and the C-terminal moieties of its cellular partners counteracts MAST2-mediated suppression of neurite outgrowth. Here, we designed peptides derived from the native viral protein to increase the affinity of these peptides for the MAST2-PDZ domain. Our strategy involved modifying the length and flexibility of the noninteracting sequence linking the two subsites anchoring the peptide to the PDZ domain. Three peptides, Neurovita1 (NV1), NV2, and NV3, were selected, and we found that they all had increased affinities for the MAST2-PDZ domain, with Kd values decreasing from 1300 to 60 nm, while target selectivity was maintained. A parallel biological assay evaluating neurite extension and branching in cell cultures revealed that the NV peptides gradually improved neural activity, with the efficacies of these peptides for stimulating neurite outgrowth mirroring their affinities for MAST2-PDZ. We also show that NVs can be delivered into the cytoplasm of neurons as a gene or peptide. In summary, our findings indicate that virus-derived peptides targeted to MAST2-PDZ stimulate neurite outgrowth in several neuron types, opening up promising avenues for potentially using NVs in the management of neurodegenerative diseases.


Asunto(s)
Neuritas/metabolismo , Proyección Neuronal/efectos de los fármacos , Dominios PDZ/fisiología , Estimulantes del Sistema Nervioso Central/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Microtúbulos/metabolismo , Neuronas/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Virus de la Rabia , Relación Estructura-Actividad , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
4.
Biologicals ; 53: 19-29, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29580693

RESUMEN

Live attenuated vaccines have proved to be mostly valuable in the prevention of infectious diseases in humans, especially in developing countries. The safety and potency of vaccine, and the consistency of vaccine batch-to-batch manufacturing, must be proven before being administrated to humans. For now, the tests used to control vaccine safety largely involve animal testing. For live viral vaccines, regulations require suppliers to demonstrate the absence of neurovirulence in animals, principally in non-human primates and mice. In a search to reduce the use of animals and embracing the 3Rs principles (Replacement, Reduction, Refinement in the use of laboratory animals), we developed a new Blood-Brain Barrier Minibrain (BBB-Minibrain) in cellulo device to evaluate the neuroinvasiveness/neurovirulence of live Yellow Fever virus (YFV) vaccines. A pilot study was performed using the features of two distinct YFV strains, with the ultimate goal of proposing a companion test to characterize YFV neurovirulence. Here, we demonstrate that the BBB-Minibrain model is a promising alternative to consider for future replacement of YFV vaccine in vivo neurovirulence testing (see graphical abstract).


Asunto(s)
Barrera Hematoencefálica/metabolismo , Modelos Inmunológicos , Vacuna contra la Fiebre Amarilla , Virus de la Fiebre Amarilla , Barrera Hematoencefálica/virología , Células Cultivadas , Humanos , Proyectos Piloto , Control de Calidad , Vacuna contra la Fiebre Amarilla/inmunología , Vacuna contra la Fiebre Amarilla/farmacocinética , Vacuna contra la Fiebre Amarilla/farmacología
5.
Plant Biotechnol J ; 15(10): 1331-1339, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28273388

RESUMEN

The biomedical applications of antibody engineering are developing rapidly and have been expanded to plant expression platforms. In this study, we have generated a novel antibody molecule in planta for targeted delivery across the blood-brain barrier (BBB). Rabies virus (RABV) is a neurotropic virus for which there is no effective treatment after entry into the central nervous system. This study investigated the use of a RABV glycoprotein peptide sequence to assist delivery of a rabies neutralizing single-chain antibody (ScFv) across an in cellulo model of human BBB. The 29 amino acid rabies virus peptide (RVG) recognizes the nicotinic acetylcholine receptor (nAchR) at neuromuscular junctions and the BBB. ScFv and ScFv-RVG fusion proteins were produced in Nicotiana benthamiana by transient expression. Both molecules were successfully expressed and purified, but the ScFv expression level was significantly higher than that of ScFv-RVG fusion. Both ScFv and ScFv-RVG fusion molecules had potent neutralization activity against RABVin cellulo. The ScFv-RVG fusion demonstrated increased binding to nAchR and entry into neuronal cells, compared to ScFv alone. Additionally, a human brain endothelial cell line BBB model was used to demonstrate that plant-produced ScFv-RVGP fusion could translocate across the cells. This study indicates that the plant-produced ScFv-RVGP fusion protein was able to cross the in celluloBBB and neutralize RABV.


Asunto(s)
Barrera Hematoencefálica , Glicoproteínas/inmunología , Fragmentos de Péptidos/inmunología , Planticuerpos/farmacología , Virus de la Rabia/inmunología , Proteínas Virales/inmunología , Anticuerpos Neutralizantes/biosíntesis , Línea Celular , Humanos , Planticuerpos/aislamiento & purificación , Planticuerpos/metabolismo , Plantas Modificadas Genéticamente , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes de Fusión , Nicotiana
6.
J Am Chem Soc ; 134(50): 20533-43, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23171049

RESUMEN

PTEN phosphatase is a tumor suppressor controlling notably cell growth, proliferation and survival. The multisite phosphorylation of the PTEN C-terminal tail regulates PTEN activity and intracellular trafficking. The dynamical nature of such regulatory events represents a crucial dimension for timing cellular decisions. Here we show that NMR spectroscopy allows reporting on the order and kinetics of clustered multisite phosphorylation events. We first unambiguously identify in vitro seven bona fide sites modified by CK2 and GSK3ß kinases and two new sites on the PTEN C-terminal tail. Then, monitoring the formation of transient intermediate phosphorylated states, we determine the sequence of these reactions and calculate their apparent rate constants. Finally, we assess the dynamic formation of these phosphorylation events induced by endogenous kinases directly in extracts of human neuroblastoma cells. Taken together, our data indicate that two cascades of events controlled by CK2 and GSK3ß occur independently on two clusters of sites (S380-S385 and S361-S370) and that in each cluster the reactions follow an ordered model with a distributive kinetic mechanism. Besides emphasizing the ability of NMR to quantitatively and dynamically follow post-translational modifications, these results bring a temporal dimension on the establishment of PTEN phosphorylation cascades.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Resonancia Magnética Nuclear Biomolecular , Fosfohidrolasa PTEN/química , Fosforilación
7.
Brain Res Bull ; 185: 107-116, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35537567

RESUMEN

Rabies is a lethal infectious disease caused by rabies virus (RABV). Fear and anxiety are the distinguished symptoms in rabies patients. Fusion of RABV envelope glycoprotein (RVG) to host cell membrane initiates rabies pathogenesis via interacting with PDZ domain of signaling proteins. We assessed the anxiety-like behaviors, and hypothalamic-pituitary-adrenal axis (HPA) response to RVG infection. Contribution of PDZ binding motif (PBM) of RVG to the observed effects was also examined using a mutant form of RVG, ΔRVG, with deleted last four amino acids at PBM C-terminus. Lentiviral vectors containing RVG and/or ΔRVG genes were injected into the rat brain areas involved in anxiety including hypothalamus, dorsal hippocampus, and amygdala. RVG/ΔRVG neural expression was examined by fluorescent microscopy. Anxiety-like behaviors were assessed by elevated plus maze (EPM) and open field (OF) tasks. HPA response was evaluated via measuring corticosterone serum level by ELISA technique. RVG/ΔRVG were successfully expressed in neurons of the injected areas. RVG, but not ΔRVG, infection of hypothalamus and amygdala increased the time spent in EPM open arms, and OF total distance moved and velocity. RVG, but not ΔRVG, infection of hypothalamus and dorsal hippocampus increased corticosterone level. The anxiety-like behaviors and exploratory/locomotor activities of rats with RVG infection in hypothalamus, and amygdala are mediated by PBM of RVG. The HPA response to RVG infection of hypothalamus and dorsal hippocampus is dependent to PBM of RVG. Triggering anxiety-related signaling by PBM of RVG seems to be one of the mechanisms involved in anxiety behaviors seen in patients with rabies.


Asunto(s)
Virus de la Rabia , Rabia , Animales , Ansiedad , Corticosterona/metabolismo , Glicoproteínas , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Virus de la Rabia/genética , Virus de la Rabia/metabolismo , Ratas
8.
PLoS Pathog ; 5(2): e1000315, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19247444

RESUMEN

Human neurons express the innate immune response receptor, Toll-like receptor 3 (TLR3). TLR3 levels are increased in pathological conditions such as brain virus infection. Here, we further investigated the production, cellular localisation, and function of neuronal TLR3 during neuronotropic rabies virus (RABV) infection in human neuronal cells. Following RABV infection, TLR3 is not only present in endosomes, as observed in the absence of infection, but also in detergent-resistant perinuclear inclusion bodies. As well as TLR3, these inclusion bodies contain the viral genome and viral proteins (N and P, but not G). The size and composition of inclusion bodies and the absence of a surrounding membrane, as shown by electron microscopy, suggest they correspond to the previously described Negri Bodies (NBs). NBs are not formed in the absence of TLR3, and TLR3(-/-) mice -- in which brain tissue was less severely infected -- had a better survival rate than WT mice. These observations demonstrate that TLR3 is a major molecule involved in the spatial arrangement of RABV-induced NBs and viral replication. This study shows how viruses can exploit cellular proteins and compartmentalisation for their own benefit.


Asunto(s)
Cuerpos de Inclusión Viral , Neuronas/virología , Virus de la Rabia/fisiología , Rabia/patología , Rabia/virología , Receptor Toll-Like 3/metabolismo , Animales , Compartimento Celular , Células Cultivadas , Interpretación Estadística de Datos , Endosomas/metabolismo , Endosomas/virología , Humanos , Cuerpos de Inclusión Viral/inmunología , Cuerpos de Inclusión Viral/metabolismo , Cuerpos de Inclusión Viral/virología , Estimación de Kaplan-Meier , Ratones , Ratones Noqueados , Microscopía Electrónica , Neuronas/metabolismo , Nucleocápside/metabolismo , Rabia/inmunología , Rabia/metabolismo , Receptor Toll-Like 3/genética , Replicación Viral
9.
NPJ Vaccines ; 6(1): 64, 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33903598

RESUMEN

Mass vaccination with the live attenuated vaccine YF-17D is the current way to prevent infection with Yellow fever virus (YFV). However, 0.000012-0.00002% of vaccinated patients develop post-vaccination neurological syndrome (YEL-AND). Understanding the factors responsible for neuroinvasion, neurotropism, and neurovirulence of the vaccine is critical for improving its biosafety. The YF-FNV vaccine strain, known to be associated with a higher frequency of YEL-AND (0.3-0.4%) than YF-17D, is an excellent model to study vaccine neuroinvasiveness. We determined that neuroinvasiveness of YF-FNV occured both via infection and passage through human brain endothelial cells. Plaque purification and next generation sequencing (NGS) identified several neuroinvasive variants. Their neuroinvasiveness was not higher than that of YF-FNV. However, rebuilding the YF-FNV population diversity from a set of isolated YF-FNV-N variants restored the original neuroinvasive phenotype of YF-FNV. Therefore, we conclude that viral population diversity is a critical factor for YFV vaccine neuroinvasiveness.

10.
PLoS One ; 16(6): e0252595, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34086776

RESUMEN

Japanese encephalitis virus (JEV) is the major cause of viral encephalitis in South East Asia. It has been suggested that, as a consequence of the inflammatory process during JEV infection, there is disruption of the blood-brain barrier (BBB) tight junctions that in turn allows the virus access to the central nervous system (CNS). However, what happens at early times of JEV contact with the BBB is poorly understood. In the present work, we evaluated the ability of both a virulent and a vaccine strain of JEV (JEV RP9 and SA14-14-2, respectively) to cross an in vitro human BBB model. Using this system, we demonstrated that both JEV RP9 and SA14-14-2 are able to cross the BBB without disrupting it at early times post viral addition. Furthermore, we find that almost 10 times more RP9 infectious particles than SA14-14 cross the model BBB, indicating this BBB model discriminates between the virulent RP9 and the vaccine SA14-14-2 strains of JEV. Beyond contributing to the understanding of early events in JEV neuroinvasion, we demonstrate this in vitro BBB model can be used as a system to study the viral determinants of JEV neuroinvasiveness and the molecular mechanisms by which this flavivirus crosses the BBB during early times of neuroinvasion.


Asunto(s)
Barrera Hematoencefálica/virología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Modelos Biológicos , Barrera Hematoencefálica/fisiología , Línea Celular , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/virología , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Virulencia , Replicación Viral
11.
J Virol ; 83(20): 10808-20, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19706704

RESUMEN

We wanted to develop a therapeutic approach against rabies disease by targeting the lyssavirus transcription/replication complex. Because this complex (nucleoprotein N-RNA template processed by the L polymerase and its cofactor, the phosphoprotein P) is similar to that of other negative-strand RNA viruses, we aimed to design broad-spectrum antiviral drugs that could be used as a complement to postexposure vaccination and immunotherapy. Recent progress in understanding the structure/function of the rabies virus P, N, and L proteins predicts that the amino-terminal end of P is an excellent target for destabilizing the replication complex because it interacts with both L (for positioning onto the N-RNA template) and N (for keeping N soluble, as needed for viral RNA encapsidation). Thus, peptides mimicking various lengths of the amino-terminal end of P have been evaluated, as follows: (i) for binding properties to the N-P-L partners by the two-hybrid method; (ii) for their capacity to inhibit the transcription/replication of a rabies virus minigenome encoding luciferase in BHK-21-T7 cells; and (iii) for their capacity to inhibit rabies virus infection of BHK-21-T7 cells and of two derivatives of the neuronal SK-N-SH cell line. Peptides P60 and P57 (the first 60 and first 57 NH2 residues of P, respectively) exhibited a rapid, strong, and long-lasting inhibitory potential on luciferase expression (>95% from 24 h to 55 h). P42 was less efficient in its inhibition level (75% for 18 to 30 h) and duration (40% after 48 h). The most promising peptides were synthesized in tandem with the Tat sequence, allowing cell penetration. Their inhibitory effects were observed on BHK-21-T7 cells infected with rabies virus and Lagos bat virus but not with vesicular stomatitis virus. In neuronal cells, a significant inhibition of both nucleocapsid inclusions and rabies virus release was observed.


Asunto(s)
Antivirales/farmacología , Péptidos/farmacología , Fosfoproteínas/química , Virus de la Rabia/efectos de los fármacos , Proteínas Estructurales Virales/química , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/química , Línea Celular , Línea Celular Tumoral , Cricetinae , Humanos , Inmunoprecipitación , Chaperonas Moleculares , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/virología , Péptidos/síntesis química , Péptidos/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Virus de la Rabia/patogenicidad , Técnicas del Sistema de Dos Híbridos , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo
12.
PLoS Negl Trop Dis ; 14(12): e0008878, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33264290

RESUMEN

Mycobacterium ulcerans, the causative agent of Buruli ulcer (BU) disease, is unique amongst human pathogens in its capacity to produce a lipid toxin called mycolactone. While previous studies have demonstrated that bacterially-released mycolactone diffuses beyond infection foci, the spatiotemporal distribution of mycolactone remained largely unknown. Here, we used the zebrafish model to provide the first global kinetic analysis of mycolactone's diffusion in vivo, and multicellular co-culture systems to address the critical question of the toxin's access to the brain. Zebrafish larvae were injected with a fluorescent-derivative of mycolactone to visualize the in vivo diffusion of the toxin from the peripheral circulation. A rapid, body-wide distribution of mycolactone was observed, with selective accumulation in tissues near the injection site and brain, together with an important excretion through the gastro-intestinal tract. Our conclusion that mycolactone reached the central nervous system was reinforced by an in cellulo model of human blood brain barrier and a mouse model of M. ulcerans-infection. Here we show that mycolactone has a broad but heterogenous profile of distribution in vivo. Our investigations in vitro and in vivo support the view that a fraction of bacterially-produced mycolactone gains access to the central nervous system. The relative persistence of mycolactone in the bloodstream suggests that assays of circulating mycolactone are relevant for BU disease monitoring and treatment optimization.


Asunto(s)
Toxinas Bacterianas/farmacocinética , Sistema Nervioso Central/metabolismo , Macrólidos/farmacocinética , Animales , Astrocitos/fisiología , Toxinas Bacterianas/administración & dosificación , Barrera Hematoencefálica , Línea Celular , Células Endoteliales/fisiología , Humanos , Larva , Macrólidos/administración & dosificación , Mycobacterium ulcerans , Imagen Óptica , Análisis Espacio-Temporal , Pez Cebra
13.
J Vis Exp ; (146)2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-31033945

RESUMEN

The early screening of nervous system medicines on a pertinent and reliable in cellulo BBB model for their penetration and their interaction with the barrier and the brain parenchyma is still an unmet need. To fill this gap, we designed a 2D in cellulo model, the BBB-Minibrain, by combining a polyester porous membrane culture insert human BBB model with a Minibrain formed by a tri-culture of human brain cells (neurons, astrocytes and microglial cells). The BBB-Minibrain allowed us to test the transport of a neuroprotective drug candidate (e.g., Neurovita), through the BBB, to determine the specific targeting of this molecule to neurons and to show that the neuroprotective property of the drug was preserved after the drug had crossed the BBB. We have also demonstrated that BBB-Minibrain constitutes an interesting model to detect the passage of virus particles across the endothelial cells barrier and to monitor the infection of the Minibrain by neuroinvasive virus particles. The BBB-Minibrain is a reliable system, easy to handle for researcher trained in cell culture technology and predictive of the brain cells phenotypes after treatment or insult. The interest of such in cellulo testing would be twofold: introducing derisking steps early in the drug development on the one hand and reducing the use of animal testing on the other hand.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Modelos Neurológicos , Fármacos Neuroprotectores/metabolismo , Animales , Astrocitos/fisiología , Barrera Hematoencefálica/fisiología , Células Cultivadas , Células Endoteliales/fisiología , Humanos , Neuronas/metabolismo , Fármacos Neuroprotectores/administración & dosificación
14.
Vaccine ; 37(33): 4673-4680, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-29523449

RESUMEN

Rabies causes more than 60,000 human deaths annually in areas where the virus is endemic. Importantly, rabies is one of the few pathogens for which there is no treatment following the onset of clinical disease with the outcome of infection being death in almost 100% of cases. Whilst vaccination, and the combination of vaccine and rabies immunoglobulin treatment for post-exposure administration are available, no tools have been identified that can reduce or prevent rabies virus replication once clinical disease has initiated. The search for effective antiviral molecules to treat those that have already developed clinical disease associated with rabies virus infection is considered one of the most important goals in rabies research. The current study assesses a single chain antibody molecule (ScFv) based on a monoclonal antibody that potently neutralises rabies in vitro as a potential therapeutic candidate. The recombinant ScFv was generated in Nicotiana benthamiana by transient expression, and was chemically conjugated (ScFv/RVG) to a 29 amino acid peptide, specific for nicotinic acetylcholine receptor (nAchR) binding in the CNS. This conjugated molecule was able to bind nAchR in vitro and enter neuronal cells more efficiently than ScFv. The ability of the ScFv/RVG to neutralise virus in vivo was assessed using a staggered administration where the molecule was inoculated either four hours before, two days after or four days after infection. The ScFv/RVG conjugate was evaluated in direct comparison with HRIG and a potential antiviral molecule, Favipiravir (also known as T-705) to indicate whether there was greater bioavailability of the ScFv in the brains of treated mice. The study indicated that the approach taken with the ScFv/RVG conjugate may have utility in the design and implementation of novel tools targetting rabies virus infection in the brain.


Asunto(s)
Vacunas Antirrábicas/uso terapéutico , Virus de la Rabia/inmunología , Rabia/metabolismo , Anticuerpos de Cadena Única/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Barrera Hematoencefálica/metabolismo , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Rabia/inmunología , Rabia/prevención & control , Vacunas Antirrábicas/inmunología , Virus de la Rabia/patogenicidad , Anticuerpos de Cadena Única/inmunología
15.
Hum Immunol ; 68(4): 294-302, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17400066

RESUMEN

Human Leukocyte Antigen (HLA)-G and E are nonclassical human MHC class I molecules. They may promote tolerance leading to virus and tumor immune escape. We recently described that the herpes simplex virus type 1 (HSV-1), a neurotropic virus inducing chronic infection and neuron latency, and rabies virus (RABV), a neuronotropic virus triggering acute neuron infection, up-regulate HLA-G expression in human neurons (NT2-N). Surface expression was only detected after RABV infection. We investigated here whether RABV and HSV-1 up-regulate HLA-E expression in human neuronal precursors (Ntera-2D/1). We found that RABV, not HSV-1, up-regulates HLA-E expression, nevertheless HLA-E could not be detected on the surface of RABV-infected Ntera-2D/1. Altogether these data suggest that HLA-G and not HLA-E could contribute to the immune escape of RABV. In contrast, there was no evidence that these molecules are used by latent HSV-1 infection. Thus, neurotropic viruses that escape the host immune response totally (RABV) or partially (HSV-1) regulate HLA-G expression on human neuronal cells differentially.


Asunto(s)
Antígenos HLA/genética , Herpesvirus Humano 1/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Neuronas/inmunología , Virus de la Rabia/inmunología , Línea Celular , Membrana Celular/inmunología , Antígenos HLA/biosíntesis , Antígenos HLA-G , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Neuronas/metabolismo , Neuronas/virología , Regulación hacia Arriba/inmunología , Antígenos HLA-E
16.
Microbes Infect ; 19(9-10): 476-484, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28602914

RESUMEN

In the field of live viral vaccines production, there is an unmet need for in vitro tests complying a 3R approach (Refine, Replace and Reduce the use of animal experimentation) to replace the post-licensing safety tests currently assayed in animals. Here, we performed a pilot study evaluating whether virulence of rabies virus, RABV, can be forecast by an in vitro test of neurite outgrowth. The rationale to use neurite outgrowth as a read-out for this test is based on the salient property of the cytoplasmic domain of the G-protein (Cyto-G) of virulent RABV strains - not of attenuated RABV strains - to stimulate neurite outgrowth in vitro. We observed that neurite elongation triggered by the Cyto-Gs encoded by different RABV field isolates correlate with the distinct virulence scores obtained in a mouse model of experimental rabies. Our results cast the idea that it could be feasible to predict RABV virulence by testing the in vitro property of a RABV strain to promote neurite outgrowth without the use of animal experimentation.


Asunto(s)
Alternativas a las Pruebas en Animales , Glicoproteínas/metabolismo , Neuritas/virología , Fragmentos de Péptidos/metabolismo , Virus de la Rabia , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Regulación Viral de la Expresión Génica/fisiología , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Neuritas/fisiología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Vacunas Antirrábicas/inmunología , Ratas , Proteínas Recombinantes de Fusión , Proteínas Virales/química , Proteínas Virales/genética
17.
Microbes Infect ; 19(9-10): 485-491, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28627433

RESUMEN

Rabies is a fatal zoonotic disease and infections generally lead to a fatal encephalomyelitis in both humans and animals. In South Africa, domestic (dogs) and the wildlife (yellow mongoose) host species maintain the canid and mongoose rabies variants respectively. In this study, pathogenicity differences of South African canid and mongoose rabies viruses were investigated in a murine model, by assessing the progression of clinical signs and survivorship. Comparison of glycoprotein gene sequences revealed amino acid differences that may underpin the observed pathogenicity differences. Cumulatively, our results suggest that the canid rabies virus may be more neurovirulent in mice than the mongoose rabies variant.


Asunto(s)
Virus de la Rabia/genética , Rabia/veterinaria , Animales , Enfermedades de los Perros/epidemiología , Enfermedades de los Perros/virología , Perros , Herpestidae , Humanos , Ratones , Filogenia , ARN Viral/genética , Rabia/epidemiología , Rabia/virología , Virus de la Rabia/patogenicidad , Sudáfrica/epidemiología , Virulencia , Zoonosis
18.
J Mol Neurosci ; 29(3): 185-94, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17085778

RESUMEN

Inflammation is an important factor in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease or multiple sclerosis, and during microbial infections of the nervous system. Glial cells were thought to be the main contributor for cytokine and chemokine production and Toll-like receptor (TLR) expression in the brain. Here, we report that human neurons express TLR-3, a major receptor in virus-mediated innate immune response. We established that these cells can mount a strong inflammatory response characterized by the expression of inflammatory cytokines (TNF-alpha, IL-6), chemokines (CCL-5 and CXCL-10), and antiviral molecules (2'5'OAS and IFN-beta) after treatment with dsRNA - a by-product of viral infection and ligand of TLR-3. This work firmly establishes that human neurons, in absence of glia, have the intrinsic machinery to trigger robust inflammatory, chemoattractive, and antiviral responses.


Asunto(s)
Encéfalo/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/metabolismo , Receptor Toll-Like 3/biosíntesis , Diferenciación Celular/efectos de los fármacos , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Quimiocinas/biosíntesis , Quimiocinas/genética , Citocinas/biosíntesis , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/inmunología , Interferón Tipo I/farmacología , Interferón beta/biosíntesis , Interferón beta/genética , Lipopolisacáridos/farmacología , Proteínas del Tejido Nervioso/genética , Neuronas/inmunología , Especificidad de Órganos , Poli I-C/farmacología , ARN Bicatenario/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 1/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética , Virosis/inmunología
19.
Ann N Y Acad Sci ; 1010: 577-81, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15033795

RESUMEN

This study sought to identify the RV protein that causes apoptosis. For this purpose, we first compared the ability of G and N proteins of a pathogenic and a nonpathogenic strain to trigger apoptosis of Jurkat rtTA by using an inducible Tet-on expression system. Then we analyzed apoptosis induced by a reverse genetic-engineered recombinant rabies virus in which the G gene from a nonpathogenic strain was replaced by its pathogenic strain counterpart. No other virus proteins than G of nonpathogenic RV strains induce apoptosis, and the G polypeptide of RV is a critical determinant for apoptosis in human cells.


Asunto(s)
Antígenos Virales , Apoptosis/fisiología , Glicoproteínas/fisiología , Virus de la Rabia/fisiología , Proteínas del Envoltorio Viral/fisiología , Apoptosis/efectos de los fármacos , Humanos , Células Jurkat , Proteínas Recombinantes/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología , Linfocitos T/virología
20.
Sci Signal ; 5(237): ra58, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22894835

RESUMEN

PTEN (phosphatase and tensin homolog deleted on chromosome 10) and MAST2 (microtubule-associated serine and threonine kinase 2) interact with each other through the PDZ domain of MAST2 (MAST2-PDZ) and the carboxyl-terminal (C-terminal) PDZ domain-binding site (PDZ-BS) of PTEN. These two proteins function as negative regulators of cell survival pathways, and silencing of either one promotes neuronal survival. In human neuroblastoma cells infected with rabies virus (RABV), the C-terminal PDZ domain of the viral glycoprotein (G protein) can target MAST2-PDZ, and RABV infection triggers neuronal survival in a PDZ-BS-dependent fashion. These findings suggest that the PTEN-MAST2 complex inhibits neuronal survival and that viral G protein disrupts this complex through competition with PTEN for binding to MAST2-PDZ. We showed that the C-terminal sequences of PTEN and the viral G protein bound to MAST2-PDZ with similar affinities. Nuclear magnetic resonance structures of these complexes exhibited similar large interaction surfaces, providing a structural basis for their binding specificities. Additionally, the viral G protein promoted the nuclear exclusion of PTEN in infected neuroblastoma cells in a PDZ-BS-dependent manner without altering total PTEN abundance. These findings suggest that formation of the PTEN-MAST2 complex is specifically affected by the viral G protein and emphasize how disruption of a critical protein-protein interaction regulates intracellular PTEN trafficking. In turn, the data show how the viral protein might be used to decipher the underlying molecular mechanisms and to clarify how the subcellular localization of PTEN regulates neuronal survival.


Asunto(s)
Glicoproteínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Neuronas/fisiología , Fosfohidrolasa PTEN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Virus de la Rabia/metabolismo , Proteínas Virales/metabolismo , Unión Competitiva , Western Blotting , Calorimetría , Línea Celular Tumoral , Supervivencia Celular/fisiología , Glicoproteínas/química , Humanos , Inmunohistoquímica , Marcaje Isotópico , Proteínas Asociadas a Microtúbulos/química , Neuronas/metabolismo , Resonancia Magnética Nuclear Biomolecular , Dominios PDZ/fisiología , Fosfohidrolasa PTEN/química , Proteínas Serina-Treonina Quinasas/química , Espectrometría de Fluorescencia , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA