Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(7): 1611-1616, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29378968

RESUMO

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.


Assuntos
Doença de Borna/virologia , Vírus da Doença de Borna/fisiologia , Transtornos Cognitivos/etiologia , Hipocampo/virologia , Memória de Longo Prazo/fisiologia , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Doença de Borna/metabolismo , Doença de Borna/patologia , Células Cultivadas , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Giro Denteado/metabolismo , Giro Denteado/patologia , Giro Denteado/virologia , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Mutação , Plasticidade Neuronal , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Fosfoproteínas/genética , Fosforilação , Proteína Quinase C/genética , Proteínas Estruturais Virais/genética
2.
PLoS Pathog ; 11(4): e1004859, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25923687

RESUMO

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.


Assuntos
Vírus da Doença de Borna/fisiologia , Regulação para Baixo , Neurônios GABAérgicos/metabolismo , Interações Hospedeiro-Patógeno , Neurogênese , Fosfoproteínas/metabolismo , Proteínas Estruturais Virais/metabolismo , Transporte Ativo do Núcleo Celular , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/metabolismo , Biomarcadores/química , Biomarcadores/metabolismo , Doença de Borna/metabolismo , Doença de Borna/patologia , Doença de Borna/virologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Proliferação de Células , Células Cultivadas , França , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/patologia , Neurônios GABAérgicos/virologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/patologia , Células-Tronco Embrionárias Humanas/virologia , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidade , Estatmina , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Estruturais Virais/genética
3.
FASEB J ; 30(4): 1523-33, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26700735

RESUMO

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.


Assuntos
Vírus da Doença de Borna/genética , Mitocôndrias/metabolismo , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Axônios/efeitos dos fármacos , Axônios/metabolismo , Western Blotting , Vírus da Doença de Borna/metabolismo , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sinais de Localização Nuclear/genética , Homologia de Sequência de Aminoácidos , Proteínas Virais/metabolismo
4.
J Virol ; 89(11): 5996-6008, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25810554

RESUMO

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.


Assuntos
Vírus da Doença de Borna/fisiologia , Epigênese Genética , Interações Hospedeiro-Patógeno , Neurônios/virologia , Fosfoproteínas/metabolismo , Proteínas Estruturais Virais/metabolismo , Replicação Viral , Acetilação , Animais , Células Cultivadas , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Ratos Sprague-Dawley
5.
J Exp Med ; 221(2)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38189779

RESUMO

The mechanisms whereby Eomes controls tissue accumulation of T cells and strengthens inflammation remain ill-defined. Here, we show that Eomes deletion in antigen-specific CD4+ T cells is sufficient to protect against central nervous system (CNS) inflammation. While Eomes is dispensable for the initial priming of CD4+ T cells, it is required for long-term maintenance of CNS-infiltrating CD4+ T cells. We reveal that the impact of Eomes on effector CD4+ T cell longevity is associated with sustained expression of multiple genes involved in mitochondrial organization and functions. Accordingly, epigenetic studies demonstrate that Eomes supports mitochondrial function by direct binding to either metabolism-associated genes or mitochondrial transcriptional modulators. Besides, the significance of these findings was confirmed in CD4+ T cells from healthy donors and multiple sclerosis patients. Together, our data reveal a new mechanism by which Eomes promotes severity and chronicity of inflammation via the enhancement of CD4+ T cell mitochondrial functions and resistance to stress-induced cell death.


Assuntos
Linfócitos T CD4-Positivos , Sistema Nervoso Central , Proteínas com Domínio T , Humanos , Morte Celular , Inflamação , Mitocôndrias , Proteínas com Domínio T/genética
6.
Neural Regen Res ; 18(2): 293-298, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35900406

RESUMO

By controlling the proper folding of proteins imported into mitochondria and ensuring crosstalk between the reticulum and mitochondria to modulate intracellular calcium fluxes, Mortalin is a chaperone protein that plays crucial roles in neuronal homeostasis and activity. However, its expression and stability are strongly modified in response to cellular stresses, in particular upon altered oxidative conditions during neurodegeneration. Here, we report and discuss the abundant literature that has highlighted its contribution to the pathophysiology of Parkinson's disease, as well as its therapeutic and prognostic potential in this still incurable pathology.

7.
Front Mol Neurosci ; 16: 1241222, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37736113

RESUMO

Dominant optic atrophy (DOA) is mainly caused by OPA1 mutations and is characterized by the degeneration of retinal ganglion cells (RGCs), whose axons form the optic nerve. The penetrance of DOA is incomplete and the disease is marked by highly variable expressivity, ranging from asymptomatic patients to some who are totally blind or who suffer from multisystemic effects. No clear genotype-phenotype correlation has been established to date. Taken together, these observations point toward the existence of modifying genetic and/or environmental factors that modulate disease severity. Here, we investigated the influence of genetic background on DOA expressivity by switching the previously described DOA mouse model bearing the c.1065 + 5G → A Opa1 mutation from mixed C3H; C57BL/6 J to a pure C57BL/6 J background. We no longer observed retinal and optic nerve abnormalities; the findings indicated no degeneration, but rather a sex-dependent negative effect on RGC connectivity. This highlights the fact that RGC synaptic alteration might precede neuronal death, as has been proposed in other neurodegenerative diseases, providing new clinical considerations for early diagnosis as well as a new therapeutic window for DOA. Furthermore, our results demonstrate the importance of secondary genetic factors in the variability of DOA expressivity and offer a model for screening for aggravating environmental and genetic factors.

8.
Sci Rep ; 11(1): 17705, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34489498

RESUMO

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.


Assuntos
Córtex Cerebral/metabolismo , Proteínas de Choque Térmico HSP70/genética , Dinâmica Mitocondrial/fisiologia , Neurônios/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Rotenona/farmacologia
9.
Neurotox Res ; 36(2): 257-267, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30215161

RESUMO

Generation of new neurons is a tightly regulated process that involves several intrinsic and extrinsic factors. Among them, a metabolic switch from glycolysis to oxidative phosphorylation, together with mitochondrial remodeling, has emerged as crucial actors of neurogenesis. However, although accumulating data raise the importance of mitochondrial morphology and function in neural stem cell proliferation and differentiation during development, information regarding the contribution of mitochondria to adult neurogenesis processes remains limited. In the present review, we discuss recent evidence covering the importance of mitochondrial morphology, function, and energy metabolism in the regulation of neuronal development and adult neurogenesis, and their impact on memory processes.


Assuntos
Mitocôndrias/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Adulto , Animais , Diferenciação Celular/fisiologia , Humanos
10.
Nat Commun ; 5: 5181, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25333748

RESUMO

Mitochondrial dysfunction is a common feature of many neurodegenerative disorders, notably Parkinson's disease. Consequently, agents that protect mitochondria have strong therapeutic potential. Here, we sought to divert the natural strategy used by Borna disease virus (BDV) to replicate in neurons without causing cell death. We show that the BDV X protein has strong axoprotective properties, thereby protecting neurons from degeneration both in tissue culture and in an animal model of Parkinson's disease, even when expressed alone outside of the viral context. We also show that intranasal administration of a cell-permeable peptide derived from the X protein is neuroprotective. We establish that both the X protein and the X-derived peptide act by buffering mitochondrial damage and inducing enhanced mitochondrial filamentation. Our results open the way to novel therapies for neurodegenerative diseases by targeting mitochondrial dynamics and thus preventing the earliest steps of neurodegenerative processes in axons.


Assuntos
Mitocôndrias/patologia , Doenças Neurodegenerativas/prevenção & controle , Doença de Parkinson/prevenção & controle , Peptídeos/química , Proteínas não Estruturais Virais/química , Animais , Axônios/metabolismo , Axônios/fisiologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Microfluídica , Microscopia Confocal , Microscopia de Fluorescência , Doenças Neurodegenerativas/virologia , Neurônios/metabolismo , Doença de Parkinson/virologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Rotenona/química
11.
PLoS One ; 9(11): e111605, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25364822

RESUMO

Bluetongue virus (BTV) is an economically important Orbivirus transmitted by biting midges to domestic and wild ruminants. The need for new vaccines has been highlighted by the occurrence of repeated outbreaks caused by different BTV serotypes since 1998. The major group-reactive antigen of BTV, VP7, is conserved in the 26 serotypes described so far, and its role in the induction of protective immunity has been proposed. Viral-based vectors as antigen delivery systems display considerable promise as veterinary vaccine candidates. In this paper we have evaluated the capacity of the BTV-2 serotype VP7 core protein expressed by either a non-replicative canine adenovirus type 2 (Cav-VP7 R0) or a leporipoxvirus (SG33-VP7), to induce immune responses in sheep. Humoral responses were elicited against VP7 in almost all animals that received the recombinant vectors. Both Cav-VP7 R0 and SG33-VP7 stimulated an antigen-specific CD4+ response and Cav-VP7 R0 stimulated substantial proliferation of antigen-specific CD8+ lymphocytes. Encouraged by the results obtained with the Cav-VP7 R0 vaccine vector, immunized animals were challenged with either the homologous BTV-2 or the heterologous BTV-8 serotype and viral burden in plasma was followed by real-time RT-PCR. The immune responses triggered by Cav-VP7 R0 were insufficient to afford protective immunity against BTV infection, despite partial protection obtained against homologous challenge. This work underscores the need to further characterize the role of BTV proteins in cross-protective immunity.


Assuntos
Antígenos Virais/genética , Vírus Bluetongue/genética , Bluetongue/imunologia , Expressão Gênica , Vetores Genéticos/genética , Proteínas do Core Viral/genética , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Bluetongue/prevenção & controle , Bluetongue/virologia , Vírus Bluetongue/imunologia , Linhagem Celular , Cricetinae , Reações Cruzadas/imunologia , Cães , Feminino , Imunidade Celular , Imunização , Masculino , Coelhos , Ovinos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas do Core Viral/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
12.
J Vis Exp ; (82): 50833, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24326926

RESUMO

Adenovirus (Ad) derived vectors have been widely used for short or long-term gene transfer, both for gene therapy and vaccine applications. Because of the frequent pre-existing immunity against the classically used human adenovirus type 5, canine adenovirus type 2 (CAV2) has been proposed as an alternative vector for human gene transfer. The well-characterized biology of CAV2, together with its ease of genetic manipulation, offer major advantages, notably for gene transfer into the central nervous system, or for inducing a wide range of protective immune responses, from humoral to cellular immunity. Nowadays, CAV2 represents one of the most appealing nonhuman adenovirus for use as a vaccine vector. This protocol describes a simple method to construct, produce and titer recombinant CAV2 vectors. After cloning the expression cassette of the gene of interest into a shuttle plasmid, the recombinant genomic plasmid is obtained by homologous recombination in the E. coli BJ5183 bacterial strain. The resulting genomic plasmid is then transfected into canine kidney cells expressing the complementing CAV2-E1 genes (DK-E1). A viral amplification enables the production of a large viral stock, which is purified by ultracentrifugation through cesium chloride gradients and desalted by dialysis. The resulting viral suspension routinely has a titer of over 10(10) infectious particles per ml and can be directly administrated in vivo.


Assuntos
Adenovirus Caninos/fisiologia , Adenovirus Caninos/genética , Adenovirus Caninos/crescimento & desenvolvimento , Animais , Linhagem Celular , Cães , Escherichia coli/genética , Vetores Genéticos/genética , Rim/citologia , Rim/virologia , Plasmídeos/genética , Transfecção , Virologia/métodos , Replicação Viral
13.
J Gen Virol ; 90(Pt 5): 1215-1223, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19264594

RESUMO

Vectors derived from canine adenovirus type 2 (CAV-2) are attractive candidates for gene therapy and live recombinant vaccines. CAV-2 vectors described thus far have been generated by modifying the virus genome, most notably early regions 1 and 3 or the fiber gene. Modification of these genes was underpinned by previous descriptions of their mRNA and protein-coding sequences. Similarly, the construction of new CAV-2 vectors bearing changes in other genomic regions, in particular many of those expressed late in the viral cycle, will require prior characterization of the corresponding transcriptional units. In this study, we provide a detailed description of the late transcriptional organization of the CAV-2 genome. We examined the major late transcription unit (MLTU) and determined its six families of mRNAs controlled by the putative major late promoter (MLP). All mRNAs expressed from the MLTU had a common non-coding tripartite leader (224 nt) at their 5' end. In transient transfection assays, the predicted MLP sequence was able to direct luciferase gene expression and the TPL sequence yielded a higher amount of transgene product. Identification of viral transcriptional products following in vitro infection confirmed most of the predicted protein-coding regions that were deduced from computer analysis of the CAV-2 genome. These findings contribute to a better understanding of gene expression in CAV-2 and lay the foundation required for genetic modifications aimed at vector optimization.


Assuntos
Adenovirus Caninos/genética , Regulação Viral da Expressão Gênica/fisiologia , Fatores de Transcrição/genética , Proteínas Virais/metabolismo , Regiões 5' não Traduzidas , Adenovirus Caninos/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Cães , Dados de Sequência Molecular , RNA Viral , Alinhamento de Sequência , Fatores de Transcrição/metabolismo , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA