Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 20(27): e2309055, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38552225

RESUMEN

Developing new approaches amenable to the measurement of neuronal physiology in real-time is a very active field of investigation, as it will offer improved methods to assess the impact of diverse insults on neuronal homeostasis. Here, the development of an in vitro bio platform is reported which can record the electrical activity of cultured primary rat cortical neurons with extreme sensitivity, while simultaneously tracking the localized changes in the pH of the culture medium. This bio platform features passive vertical nanoprobes with ultra-high signal resolution (several mV amplitude ranges) and Chem-FinFETs (pH sensitivity of sub-0.1 pH units), covering an area as little as a neuronal soma. These multi-sensing units are arranged in an array to probe both chemically and electrically an equivalent surface of ≈ 0.5 mm2. A homemade setup is also developed which allows recording of multiplexed data in real-time (10 ps range) from the active chem-sensors and passive electrodes and which is used to operate the platform. Finally, a proof-of-concept is presented for a neuro-relevant application, by investigating the effect on neuronal activity of Amyloid beta oligomers, the main toxic peptide in Alzheimer's Disease, which reveals that exposure to amyloid beta oligomers modify the amplitude, but not the frequency, of neuronal firing, without any detectable changes in pH values along this process.


Asunto(s)
Neuronas , Concentración de Iones de Hidrógeno , Neuronas/fisiología , Animales , Ratas , Electrodos , Péptidos beta-Amiloides/química , Células Cultivadas
2.
J Pathol ; 254(1): 92-102, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33565082

RESUMEN

Congenital infection of the central nervous system by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae, including mental retardation or neurodevelopmental abnormalities. The most severe complications include smooth brain or polymicrogyria, which are both indicative of abnormal migration of neural cells, although the underlying mechanisms remain to be determined. To gain better insight on the pathogenesis of such sequelae, we assessed the expression levels of a set of neurogenesis-related genes, using HCMV-infected human neural stem cells derived from embryonic stem cells (NSCs). Among the 84 genes tested, we found dramatically increased expression of the gene PAFAH1B1, encoding LIS1 (lissencephaly-1), in HCMV-infected versus uninfected NSCs. Consistent with these findings, western blotting and immunofluorescence analyses confirmed the increased levels of LIS1 in HCMV-infected NSCs at the protein level. We next assessed the migratory abilities of HCMV-infected NSCs and observed that infection strongly impaired the migration of NSCs, without detectable effect on their proliferation. Moreover, we observed increased immunostaining for LIS1 in brains of congenitally infected fetuses, but not in control samples, highlighting the clinical relevance of our findings. Of note, PAFAH1B1 mutations (resulting in either haploinsufficiency or gain of function) are primary causes of hereditary neurodevelopmental diseases. Notably, mutations resulting in PAFAH1B1 haploinsufficiency cause classic lissencephaly. Taken together, our findings suggest that PAFAH1B1 is a critical target of HCMV infection. They also shine a new light on the pathophysiological basis of the neurological outcomes of congenital HCMV infection, by suggesting that defective neural cell migration might contribute to the pathogenesis of the neurodevelopmental sequelae of infection. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/virología , Encéfalo/metabolismo , Encéfalo/virología , Infecciones por Citomegalovirus/complicaciones , Humanos
3.
Virologie (Montrouge) ; 26(4): 275-281, 2022 07 01.
Artículo en Francés | MEDLINE | ID: mdl-36120974

RESUMEN

The reality of human infections by Bornaviridae (and particularly by mammalian Orthobornaviruses BoDV-1 and BoDV-2) has long been the centre of debate and controversies. New data, however, have profoundly modified the game by providing strong and unambiguous pieces of evidence, even if many points still need to be clarified. This review aims at presenting the current state of the question, based on today's knowledge.


La question de la réalité des infections humaines par les Bornaviridae (et plus précisément par les Orthobornavirus des mammifères BoDV-1 ou BoDV-2) a longtemps constitué un point de controverse. Des données récentes ont cependant profondément remanié les cartes et permettent désormais d'avoir quelques données solides en la matière. Il n'en reste pas moins que plusieurs aspects restent mal compris. Cette revue vise à faire le point, au vu de nos connaissances à ce jour.


Asunto(s)
Bornaviridae , Animales , Bornaviridae/genética , Humanos , Mamíferos , ARN Viral
4.
Proc Natl Acad Sci U S A ; 115(7): 1611-1616, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29378968

RESUMEN

The analysis of the biology of neurotropic viruses, notably of their interference with cellular signaling, provides a useful tool to get further insight into the role of specific pathways in the control of behavioral functions. Here, we exploited the natural property of a viral protein identified as a major effector of behavioral disorders during infection. We used the phosphoprotein (P) of Borna disease virus, which acts as a decoy substrate for protein kinase C (PKC) when expressed in neurons and disrupts synaptic plasticity. By a lentiviral-based strategy, we directed the singled-out expression of P in the dentate gyrus of the hippocampus and we examined its impact on mouse behavior. Mice expressing the P protein displayed increased anxiety and impaired long-term memory in contextual and spatial memory tasks. Interestingly, these effects were dependent on P protein phosphorylation by PKC, as expression of a mutant form of P devoid of its PKC phosphorylation sites had no effect on these behaviors. We also revealed features of behavioral impairment induced by P protein expression but that were independent of its phosphorylation by PKC. Altogether, our findings provide insight into the behavioral correlates of viral infection, as well as into the impact of virus-mediated alterations of the PKC pathway on behavioral functions.


Asunto(s)
Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/fisiología , Trastornos del Conocimiento/etiología , Hipocampo/virología , Memoria a Largo Plazo/fisiología , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Enfermedad de Borna/metabolismo , Enfermedad de Borna/patología , Células Cultivadas , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Giro Dentado/metabolismo , Giro Dentado/patología , Giro Dentado/virología , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Mutación , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Fosfoproteínas/genética , Fosforilación , Proteína Quinasa C/genética , Proteínas Estructurales Virales/genética
5.
PLoS Pathog ; 12(4): e1005547, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27078877

RESUMEN

Congenital infection by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae of the central nervous system, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities (0.1% of all births). To gain insight on the impact of HCMV on neuronal development, we used both neural stem cells from human embryonic stem cells (NSC) and brain sections from infected fetuses and investigated the outcomes of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor critical in the developing brain. We observed that HCMV infection dramatically impaired the rate of neuronogenesis and strongly increased PPARγ levels and activity. Consistent with these findings, levels of 9-hydroxyoctadecadienoic acid (9-HODE), a known PPARγ agonist, were significantly increased in infected NSCs. Likewise, exposure of uninfected NSCs to 9-HODE recapitulated the effect of infection on PPARγ activity. It also increased the rate of cells expressing the IE antigen in HCMV-infected NSCs. Further, we demonstrated that (1) pharmacological activation of ectopically expressed PPARγ was sufficient to induce impaired neuronogenesis of uninfected NSCs, (2) treatment of uninfected NSCs with 9-HODE impaired NSC differentiation and (3) treatment of HCMV-infected NSCs with the PPARγ inhibitor T0070907 restored a normal rate of differentiation. The role of PPARγ in the disease phenotype was strongly supported by the immunodetection of nuclear PPARγ in brain germinative zones of congenitally infected fetuses (N = 20), but not in control samples. Altogether, our findings reveal a key role for PPARγ in neurogenesis and in the pathophysiology of HCMV congenital infection. They also pave the way to the identification of PPARγ gene targets in the infected brain.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/metabolismo , Células-Madre Neurales/virología , Neurogénesis/fisiología , PPAR gamma/metabolismo , Western Blotting , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Cromatografía Líquida de Alta Presión , Técnica del Anticuerpo Fluorescente , Humanos , Microscopía Electrónica de Transmisión , Células-Madre Neurales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem
6.
Langmuir ; 34(22): 6612-6620, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29754481

RESUMEN

Despite significant progress, our knowledge of the functioning of the central nervous system still remains scarce to date. A better understanding of its behavior, in either normal or diseased conditions, goes through an increased knowledge of basic mechanisms involved in neuronal function, including at the single-cell level. This has motivated significant efforts for the development of miniaturized sensing devices to monitor neuronal activity with high spatial and signal resolution. One of the main challenges remaining to be addressed in this domain is, however, the ability to create in vitro spatially ordered neuronal networks at low density with a precise control of the cell location to ensure proper monitoring of the activity of a defined set of neurons. Here, we present a novel self-aligned chemical functionalization method, based on a repellant surface with patterned attractive areas, which permits the elaboration of low-density neuronal network down to individual cells with a high control of the soma location and axonal growth. This approach is compatible with complementary metal-oxide-semiconductor line technology at a wafer scale and allows performing the cell culture on packaged chip outside microelectronics facilities. Rat cortical neurons were cultured on such patterned surfaces for over one month and displayed a very high degree of organization in large networks. Indeed, more than 90% of the network nodes were settled by a soma and 100% of the connecting lines were occupied by a neurite, with a very good selectivity (low parasitic cell connections). After optimization, networks composed of 75% of unicellular nodes were obtained, together with a control at the micron scale of the location of the somas. Finally, we demonstrated that the dendritic neuronal growth was guided by the surface functionalization, even when micrometer scale topologies were encountered and we succeeded to control the extension growth along one-dimensional-aligned nanostructures with sub-micrometrical scale precision. This novel approach now opens the way for precise monitoring of neuronal network activity at the single-cell level.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Red Nerviosa/química , Animales , Células Cultivadas , Dendritas , Red Nerviosa/metabolismo , Neuritas , Neuronas/citología , Ratas
7.
PLoS Pathog ; 11(4): e1004859, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25923687

RESUMEN

It is well established that persistent viral infection may impair cellular function of specialized cells without overt damage. This concept, when applied to neurotropic viruses, may help to understand certain neurologic and neuropsychiatric diseases. Borna disease virus (BDV) is an excellent example of a persistent virus that targets the brain, impairs neural functions without cell lysis, and ultimately results in neurobehavioral disturbances. Recently, we have shown that BDV infects human neural progenitor cells (hNPCs) and impairs neurogenesis, revealing a new mechanism by which BDV may interfere with brain function. Here, we sought to identify the viral proteins and molecular pathways that are involved. Using lentiviral vectors for expression of the bdv-p and bdv-x viral genes, we demonstrate that the phosphoprotein P, but not the X protein, diminishes human neurogenesis and, more particularly, GABAergic neurogenesis. We further reveal a decrease in pro-neuronal factors known to be involved in neuronal differentiation (ApoE, Noggin, TH and Scg10/Stathmin2), demonstrating that cellular dysfunction is associated with impairment of specific components of the molecular program that controls neurogenesis. Our findings thus provide the first evidence that a viral protein impairs GABAergic human neurogenesis, a process that is dysregulated in several neuropsychiatric disorders. They improve our understanding of the mechanisms by which a persistent virus may interfere with brain development and function in the adult.


Asunto(s)
Virus de la Enfermedad de Borna/fisiología , Regulación hacia Abajo , Neuronas GABAérgicas/metabolismo , Interacciones Huésped-Patógeno , Neurogénesis , Fosfoproteínas/metabolismo , Proteínas Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Biomarcadores/química , Biomarcadores/metabolismo , Enfermedad de Borna/metabolismo , Enfermedad de Borna/patología , Enfermedad de Borna/virología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proliferación Celular , Células Cultivadas , Francia , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/patología , Neuronas GABAérgicas/virología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Células Madre Embrionarias Humanas/virología , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Estatmina , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/metabolismo , Proteínas Estructurales Virales/genética
8.
FASEB J ; 30(4): 1523-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26700735

RESUMEN

To favor their replication, viruses express proteins that target diverse mammalian cellular pathways. Due to the limited size of many viral genomes, such proteins are endowed with multiple functions, which require targeting to different subcellular compartments. One salient example is the X protein of Borna disease virus, which is expressed both at the mitochondria and in the nucleus. Moreover, we recently demonstrated that mitochondrial X protein is neuroprotective. In this study, we sought to examine the mechanisms whereby the X protein transits between subcellular compartments and to define its localization signals, to enhance its mitochondrial accumulation and thus, potentially, its neuroprotective activity. We transfected plasmids expressing fusion proteins bearing different domains of X fused to enhanced green fluorescent protein (eGFP) and compared their subcellular localization to that of eGFP. We observed that the 5-16 domain of X was responsible for both nuclear export and mitochondrial targeting and identified critical residues for mitochondrial localization. We next took advantage of these findings and constructed mutant X proteins that were targeted only to the mitochondria. Such mutants exhibited enhanced neuroprotective properties in compartmented cultures of neurons grown in microfluidic chambers, thereby confirming the parallel between mitochondrial accumulation of the X protein and its neuroprotective potential.-Ferré C. A., Davezac, N., Thouard, A., Peyrin, J. M., Belenguer, P., Miquel, M.-C., Gonzalez-Dunia, D., Szelechowski, M. Manipulation of the N-terminal sequence of the Borna disease virus X protein improves its mitochondrial targeting and neuroprotective potential.


Asunto(s)
Virus de la Enfermedad de Borna/genética , Mitocondrias/metabolismo , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Axones/efectos de los fármacos , Axones/metabolismo , Western Blotting , Virus de la Enfermedad de Borna/metabolismo , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Señales de Localización Nuclear/genética , Homología de Secuencia de Aminoácido , Proteínas Virales/metabolismo
9.
J Gen Virol ; 97(12): 3215-3224, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27902378

RESUMEN

Long-range axonal retrograde transport is a key mechanism for the cellular dissemination of neuroinvasive viruses, such as Borna disease virus (BDV), for which entry and egress sites are usually distant from the nucleus, where viral replication takes place. Although BDV is known to disseminate very efficiently in neurons, both in vivo and in primary cultures, the modalities of its axonal transport are still poorly characterized. In this work, we combined different methodological approaches, such as confocal microscopy and biochemical purification of endosomes, to study BDV retrograde transport. We demonstrate that BDV ribonucleoparticles (composed of the viral genomic RNA, nucleoprotein and phosphoprotein), as well as the matrix protein, are transported towards the nucleus into endocytic carriers. These specialized organelles, called signalling endosomes, are notably used for the retrograde transport of neurotrophins and activated growth factor receptors. Signalling endosomes have a neutral luminal pH and thereby offer protection against degradation during long-range transport. This particularity could allow the viral particles to be delivered intact to the cell body of neurons, avoiding their premature release in the cytoplasm.


Asunto(s)
Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/metabolismo , Endosomas/virología , Neuronas/virología , Animales , Enfermedad de Borna/metabolismo , Virus de la Enfermedad de Borna/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Endosomas/metabolismo , Neuronas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virión/genética , Virión/metabolismo
10.
J Virol ; 89(11): 5996-6008, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810554

RESUMEN

UNLABELLED: Understanding the modalities of interaction of neurotropic viruses with their target cells represents a major challenge that may improve our knowledge of many human neurological disorders for which viral origin is suspected. Borna disease virus (BDV) represents an ideal model to analyze the molecular mechanisms of viral persistence in neurons and its consequences for neuronal homeostasis. It is now established that BDV ensures its long-term maintenance in infected cells through a stable interaction of viral components with the host cell chromatin, in particular, with core histones. This has led to our hypothesis that such an interaction may trigger epigenetic changes in the host cell. Here, we focused on histone acetylation, which plays key roles in epigenetic regulation of gene expression, notably for neurons. We performed a comparative analysis of histone acetylation patterns of neurons infected or not infected by BDV, which revealed that infection decreases histone acetylation on selected lysine residues. We showed that the BDV phosphoprotein (P) is responsible for these perturbations, even when it is expressed alone independently of the viral context, and that this action depends on its phosphorylation by protein kinase C. We also demonstrated that BDV P inhibits cellular histone acetyltransferase activities. Finally, by pharmacologically manipulating cellular acetylation levels, we observed that inhibiting cellular acetyl transferases reduces viral replication in cell culture. Our findings reveal that manipulation of cellular epigenetics by BDV could be a means to modulate viral replication and thus illustrate a fascinating example of virus-host cell interaction. IMPORTANCE: Persistent DNA viruses often subvert the mechanisms that regulate cellular chromatin dynamics, thereby benefitting from the resulting epigenetic changes to create a favorable milieu for their latent and persistent states. Here, we reasoned that Borna disease virus (BDV), the only RNA virus known to durably persist in the nucleus of infected cells, notably neurons, might employ a similar mechanism. In this study, we uncovered a novel modality of virus-cell interaction in which BDV phosphoprotein inhibits cellular histone acetylation by interfering with histone acetyltransferase activities. Manipulation of cellular histone acetylation is accompanied by a modulation of viral replication, revealing a perfect adaptation of this "ancient" virus to its host that may favor neuronal persistence and limit cellular damage.


Asunto(s)
Virus de la Enfermedad de Borna/fisiología , Epigénesis Genética , Interacciones Huésped-Patógeno , Neuronas/virología , Fosfoproteínas/metabolismo , Proteínas Estructurales Virales/metabolismo , Replicación Viral , Acetilación , Animales , Células Cultivadas , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Ratas Sprague-Dawley
12.
J Virol ; 87(22): 12339-48, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24027309

RESUMEN

Borna disease virus (BDV) is a nonsegmented, negative-stranded RNA virus characterized by noncytolytic persistent infection and replication in the nuclei of infected cells. To gain further insight on the intracellular trafficking of BDV components during infection, we sought to generate recombinant BDV (rBDV) encoding fluorescent fusion viral proteins. We successfully rescued a virus bearing a tetracysteine tag fused to BDV-P protein, which allowed assessment of the intracellular distribution and dynamics of BDV using real-time live imaging. In persistently infected cells, viral nuclear inclusions, representing viral factories tethered to chromatin, appeared to be extremely static and stable, contrasting with a very rapid and active trafficking of BDV components in the cytoplasm. Photobleaching (fluorescence recovery after photobleaching [FRAP] and fluorescence loss in photobleaching [FLIP]) imaging approaches revealed that BDV components were permanently and actively exchanged between cellular compartments, including within viral inclusions, albeit with a fraction of BDV-P protein not mobile in these structures, presumably due to its association with viral and/or cellular proteins. We also obtained evidence for transfer of viral material between persistently infected cells, with routing of the transferred components toward the cell nucleus. Finally, coculture experiments with noninfected cells allowed visualization of cell-to-cell BDV transmission and movement of the incoming viral material toward the nucleus. Our data demonstrate the potential of tetracysteine-tagged recombinant BDV for virus tracking during infection, which may provide novel information on the BDV life cycle and on the modalities of its interaction with the nuclear environment during viral persistence.


Asunto(s)
Enfermedad de Borna/virología , Virus de la Enfermedad de Borna/patogenicidad , Núcleo Celular/metabolismo , Cisteína/química , Citoplasma/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virales de Fusión/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Northern Blotting , Western Blotting , Enfermedad de Borna/metabolismo , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Fosfoproteínas/genética , Transporte de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero , Proteínas Virales de Fusión/genética , Proteínas Estructurales Virales/genética
13.
Virologie (Montrouge) ; 18(4): 187-200, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065931

RESUMEN

Viruses have to meet the challenge to cope with the limited capacity of renewal of neuronal cells in order to allow their replication and persistence in the central nervous system (CNS). Accordingly, many neurotropic viruses establish latency to optimize their maintenance in the CNS. Bornaviruses have evolved a different and original strategy to persist in neurons, which involves an active replication without associated cytopathic effect. Despite their small genomes and limited number of proteins, bornaviruses hijack multiple signaling pathways, leading to escape from immune surveillance or protection of cells against apoptosis. Long term persistence has even led to integration of genome elements within the host cell genome, leading to "fossil bornaviruses" in a wide range of vertebrate species. Hence, bornaviruses represent the ideal host-cell adaptation example and can thus be considered as the "best enemy" for its hosts.

14.
PLoS Pathog ; 7(11): e1002393, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22114563

RESUMEN

Following infection of the central nervous system (CNS), the immune system is faced with the challenge of eliminating the pathogen without causing significant damage to neurons, which have limited capacities of renewal. In particular, it was thought that neurons were protected from direct attack by cytotoxic T lymphocytes (CTL) because they do not express major histocompatibility class I (MHC I) molecules, at least at steady state. To date, most of our current knowledge on the specifics of neuron-CTL interaction is based on studies artificially inducing MHC I expression on neurons, loading them with exogenous peptide and applying CTL clones or lines often differentiated in culture. Thus, much remains to be uncovered regarding the modalities of the interaction between infected neurons and antiviral CD8 T cells in the course of a natural disease. Here, we used the model of neuroinflammation caused by neurotropic Borna disease virus (BDV), in which virus-specific CTL have been demonstrated as the main immune effectors triggering disease. We tested the pathogenic properties of brain-isolated CD8 T cells against pure neuronal cultures infected with BDV. We observed that BDV infection of cortical neurons triggered a significant up regulation of MHC I molecules, rendering them susceptible to recognition by antiviral CTL, freshly isolated from the brains of acutely infected rats. Using real-time imaging, we analyzed the spatio-temporal relationships between neurons and CTL. Brain-isolated CTL exhibited a reduced mobility and established stable contacts with BDV-infected neurons, in an antigen- and MHC-dependent manner. This interaction induced rapid morphological changes of the neurons, without immediate killing or impairment of electrical activity. Early signs of neuronal apoptosis were detected only hours after this initial contact. Thus, our results show that infected neurons can be recognized efficiently by brain-isolated antiviral CD8 T cells and uncover the unusual modalities of CTL-induced neuronal damage.


Asunto(s)
Enfermedad de Borna/inmunología , Virus de la Enfermedad de Borna/inmunología , Neuronas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Histocompatibilidad Clase I/biosíntesis , Neuronas/patología , Neuronas/virología , Ratas , Ratas Endogámicas Lew
15.
iScience ; 25(1): 103621, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35024577

RESUMEN

Borna disease viruses (BoDV) have recently emerged as zoonotic neurotropic pathogens. These persistent RNA viruses assemble nuclear replication centers (vSPOT) in close interaction with the host chromatin. However, the topology of this interaction and its consequences on neuronal function remain unexplored. In neurons, DNA double-strand breaks (DSB) have been identified as novel epigenetic mechanisms regulating neurotransmission and cognition. Activity-dependent DSB contribute critically to neuronal plasticity processes, which could be impaired upon infection. Here, we show that BoDV-1 infection, or the singled-out expression of viral Nucleoprotein and Phosphoprotein, increases neuronal DSB levels. Of interest, inducing DSB promoted the recruitment anew of vSPOT colocalized with DSB and increased viral RNA replication. BoDV-1 persistence decreased neuronal activity and response to stimulation by dampening the surface expression of glutamate receptors. Taken together, our results propose an original mechanistic cross talk between persistence of an RNA virus and neuronal function, through the control of DSB levels.

16.
Viruses ; 14(11)2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36366462

RESUMEN

Determining the structural organisation of viral replication complexes and unravelling the impact of infection on cellular homeostasis represent important challenges in virology. This may prove particularly useful when confronted with viruses that pose a significant threat to human health, that appear unique within their family, or for which knowledge is scarce. Among Mononegavirales, bornaviruses (family Bornaviridae) stand out due to their compact genomes and their nuclear localisation for replication. The recent recognition of the zoonotic potential of several orthobornaviruses has sparked a surge of interest in improving our knowledge on this viral family. In this work, we provide a complete analysis of the structural organisation of Borna disease virus 1 (BoDV-1) phosphoprotein (P), an important cofactor for polymerase activity. Using X-ray diffusion and diffraction experiments, we revealed that BoDV-1 P adopts a long coiled-coil α-helical structure split into two parts by an original ß-strand twist motif, which is highly conserved across the members of whole Orthobornavirus genus and may regulate viral replication. In parallel, we used BioID to determine the proximal interactome of P in living cells. We confirmed previously known interactors and identified novel proteins linked to several biological processes such as DNA repair or mRNA metabolism. Altogether, our study provides important structure/function cues, which may improve our understanding of BoDV-1 pathogenesis.


Asunto(s)
Virus de la Enfermedad de Borna , Bornaviridae , Animales , Humanos , Virus de la Enfermedad de Borna/genética , Fosfoproteínas/genética , Bornaviridae/genética , Reparación del ADN , ADN , ARN Mensajero/genética
17.
PLoS Pathog ; 5(5): e1000425, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19424436

RESUMEN

Understanding the pathogenesis of infection by neurotropic viruses represents a major challenge and may improve our knowledge of many human neurological diseases for which viruses are thought to play a role. Borna disease virus (BDV) represents an attractive model system to analyze the molecular mechanisms whereby a virus can persist in the central nervous system (CNS) and lead to altered brain function, in the absence of overt cytolysis or inflammation. Recently, we showed that BDV selectively impairs neuronal plasticity through interfering with protein kinase C (PKC)-dependent signaling in neurons. Here, we tested the hypothesis that BDV phosphoprotein (P) may serve as a PKC decoy substrate when expressed in neurons, resulting in an interference with PKC-dependent signaling and impaired neuronal activity. By using a recombinant BDV with mutated PKC phosphorylation site on P, we demonstrate the central role of this protein in BDV pathogenesis. We first showed that the kinetics of dissemination of this recombinant virus was strongly delayed, suggesting that phosphorylation of P by PKC is required for optimal viral spread in neurons. Moreover, neurons infected with this mutant virus exhibited a normal pattern of phosphorylation of the PKC endogenous substrates MARCKS and SNAP-25. Finally, activity-dependent modulation of synaptic activity was restored, as assessed by measuring calcium dynamics in response to depolarization and the electrical properties of neuronal networks grown on microelectrode arrays. Therefore, preventing P phosphorylation by PKC abolishes viral interference with neuronal activity in response to stimulation. Our findings illustrate a novel example of viral interference with a differentiated neuronal function, mainly through competition with the PKC signaling pathway. In addition, we provide the first evidence that a viral protein can specifically interfere with stimulus-induced synaptic plasticity in neurons.


Asunto(s)
Virus de la Enfermedad de Borna/fisiología , Neuronas/virología , Proteína Quinasa C/genética , Sinapsis/virología , Transmisión Sináptica/fisiología , Potenciales de Acción , Animales , Western Blotting , Enfermedad de Borna/fisiopatología , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Mutación , Plasticidad Neuronal/fisiología , Técnicas de Placa-Clamp , Fosfoproteínas , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
Sci Rep ; 11(1): 17705, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489498

RESUMEN

Mortalin is a mitochondrial chaperone protein involved in quality control of proteins imported into the mitochondrial matrix, which was recently described as a sensor of neuronal stress. Mortalin is down-regulated in neurons of patients with neurodegenerative diseases and levels of Mortalin expression are correlated with neuronal fate in animal models of Alzheimer's disease or cerebral ischemia. To date, however, the links between Mortalin levels, its impact on mitochondrial function and morphology and, ultimately, the initiation of neurodegeneration, are still unclear. In the present study, we used lentiviral vectors to over- or under-express Mortalin in primary neuronal cultures. We first analyzed the early events of neurodegeneration in the axonal compartment, using oriented neuronal cultures grown in microfluidic-based devices. We observed that Mortalin down-regulation induced mitochondrial fragmentation and axonal damage, whereas its over-expression conferred protection against axonal degeneration mediated by rotenone exposure. We next demonstrated that Mortalin levels modulated mitochondrial morphology by acting on DRP1 phosphorylation, thereby further illustrating the crucial implication of mitochondrial dynamics on neuronal fate in degenerative diseases.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Dinámicas Mitocondriales/fisiología , Neuronas/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Rotenona/farmacología
19.
Front Cell Dev Biol ; 9: 689122, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34568315

RESUMEN

Extracellular vesicles (EVs) have increasingly been recognized as key players in a wide variety of physiological and pathological contexts, including during pregnancy. Notably, EVs appear both as possible biomarkers and as mediators involved in the communication of the placenta with the maternal and fetal sides. A better understanding of the physiological and pathological roles of EVs strongly depends on the development of adequate and reliable study models, specifically at the beginning of pregnancy where many adverse pregnancy outcomes have their origin. In this study, we describe the isolation of small EVs from a histoculture model of first trimester placental explants in normal conditions as well as upon infection by human cytomegalovirus. Using bead-based multiplex cytometry and electron microscopy combined with biochemical approaches, we characterized these small EVs and defined their associated markers and ultrastructure. We observed that infection led to changes in the expression level of several surface markers, without affecting the secretion and integrity of small EVs. Our findings lay the foundation for studying the functional role of EVs during early pregnancy, along with the identification of new predictive biomarkers for the severity and outcome of this congenital infection, which are still sorely lacking.

20.
Arch Virol ; 155(5): 789-93, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20333534

RESUMEN

Mutational analysis of the phosphate acceptor sites of the Borna disease virus (BDV) phosphoprotein (P) has suggested a role of phosphorylation for viral spread. However, the studied mutant viruses also had two amino acid exchanges in the X protein, because the reading frames of P and X overlap. To determine the relative contribution of P and X to viral attenuation, we studied a P variant with serine-to-leucine substitutions (P(S26L,S28L)) in which the wild-type X sequence was conserved. Viral spread of rBDV-P(S26L,S28L) was impaired in human oligodendroglioma cells and in adult rats. Thus, BDV-P phosphorylation contributes to efficient viral dissemination.


Asunto(s)
Virus de la Enfermedad de Borna/fisiología , Fosfoproteínas/metabolismo , Proteína Quinasa C/fisiología , Proteínas Estructurales Virales/metabolismo , Animales , Células Cultivadas , Humanos , Fosforilación , Proteína Quinasa C/química , Ratas , Ratas Endogámicas Lew
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA