Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Biomed J ; : 100659, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37690583

ABSTRACT

The role of extracellular vesicles (EVs) as key players in the intercellular communication is a subject of growing interest in all areas of physiology and pathophysiology, and the field of viral infections is no exception to the rule. In this review, we focus on the current state of knowledge and remaining gaps regarding the entanglement of viruses and EVs during infections. These two entities share many similarities, mainly due to their intricated biogenesis pathways that are in constant interaction. EVs can promote the replication and dissemination of viruses within the organism, through the dysregulation of their cargo and the modulation of the innate and adaptive immune response that occurs upon infection, but they can also promote the mitigation of viral infections. Here, we will examine how viruses hijack EV biogenesis pathways and describe the consequences of dysregulated EV secretion during viral infections, beneficial or not for viruses, revealing the duality of their possible effects.

3.
Biochem Soc Trans ; 50(6): 1785-1795, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36484632

ABSTRACT

For several years, a growing number of studies have highlighted the pivotal role of placental extracellular vesicles (EVs) throughout pregnancy. These membrane nanovesicles, heterogeneous in nature, composition and origin, are secreted by several trophoblastic cell types and are found in both the maternal and fetal compartments. They can be uptaken by recipient cells and drive a wide variety of physiological and pathological processes. In this review, we provide an overview of the different described roles of placental EVs in various aspects of normal pregnancy, from placenta establishment to maternal immune tolerance towards the fetus and protection against viral infections. In the second part, we present selected examples of pathological pregnancies in which placental EVs are involved, such as gestational diabetes mellitus, pre-eclampsia, and congenital infections. Since the abundance and/or composition of placental EVs is deregulated in maternal serum during pathological pregnancies, this makes them interesting candidates as non-invasive biomarkers for gestational diseases and opens a wide field of translational perspectives.


Subject(s)
Extracellular Vesicles , Pre-Eclampsia , Pregnancy , Female , Humans , Placenta/metabolism , Extracellular Vesicles/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Trophoblasts/metabolism , Communication
4.
Viruses ; 14(9)2022 09 13.
Article in English | MEDLINE | ID: mdl-36146834

ABSTRACT

Although placental small extracellular vesicles (sEVs) are extensively studied in the context of pregnancy, little is known about their role during viral congenital infection, especially at the beginning of pregnancy. In this study, we examined the consequences of human cytomegalovirus (hCMV) infection on sEVs production, composition, and function using an immortalized human cytotrophoblast cell line derived from first trimester placenta. By combining complementary approaches of biochemistry, electron microscopy, and quantitative proteomic analysis, we showed that hCMV infection increases the yield of sEVs produced by cytotrophoblasts and modifies their protein content towards a potential proviral phenotype. We further demonstrate that sEVs secreted by hCMV-infected cytotrophoblasts potentiate infection in naive recipient cells of fetal origin, including human neural stem cells. Importantly, these functional consequences are also observed with sEVs prepared from an ex vivo model of infected histocultures from early placenta. Based on these findings, we propose that placental sEVs could be important actors favoring viral dissemination to the fetal brain during hCMV congenital infection.


Subject(s)
Cytomegalovirus Infections , Extracellular Vesicles , Cytomegalovirus/genetics , Extracellular Vesicles/metabolism , Female , Humans , Placenta , Pregnancy , Proteomics
5.
Viruses ; 14(8)2022 07 25.
Article in English | MEDLINE | ID: mdl-35893684

ABSTRACT

Usutu virus (USUV) is a neurotropic mosquito-borne flavivirus that has dispersed quickly in Europe these past years. This arbovirus mainly follows an enzootic cycle involving mosquitoes and birds, but can also infect other mammals, causing notably sporadic cases in humans. Although it is mainly asymptomatic or responsible for mild clinical symptoms, USUV has been associated with neurological disorders, such as encephalitis and meningoencephalitis, highlighting the potential health threat of this virus. Among the different transmission routes described for other flaviviruses, the capacity for some of them to be transmitted vertically has been demonstrated, notably for Zika virus or West Nile virus, which are closely related to USUV. To evaluate the ability of USUV to replicate in the placenta and gain access to the fetus, we combined the use of several trophoblast model cell lines, ex vivo human placental explant cultures from first and third trimester of pregnancy, and in vivo USUV-infected pregnant mice. Our data demonstrate that human placental cells and tissues are permissive to USUV replication, and suggest that viral transmission can occur in mice during gestation. Hence, our observations suggest that USUV could be efficiently transmitted by the vertical route.


Subject(s)
Culicidae , Flavivirus Infections , Flavivirus , West Nile virus , Zika Virus Infection , Zika Virus , Animals , Female , Humans , Mice , Placenta , Pregnancy
6.
Front Cell Dev Biol ; 9: 689122, 2021.
Article in English | MEDLINE | ID: mdl-34568315

ABSTRACT

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.

7.
Acta Physiol (Oxf) ; 229(1): e13440, 2020 05.
Article in English | MEDLINE | ID: mdl-31925934

ABSTRACT

AIM: Astroglial connexins (Cxs) 30 and 43 are engaged in gap junction and hemichannel activities. Evidence suggests that these functional entities contribute to regulating neurotransmission, thereby influencing brain functions. In particular, preclinical and clinical findings highlight a role of Cx43 in animal models of depression. However, the role of these proteins in response to currently available psychotropic drugs is still unknown. METHODS: To investigate this, we evaluated the behavioural effects of the genetic and pharmacological inactivation of Cx43 on the antidepressant- and anxiolytic-like activities of the selective serotonin reuptake inhibitor fluoxetine and the benzodiazepine diazepam, respectively. RESULTS: A single administration of fluoxetine (18 mg/kg; i.p.) produced a higher increase in hippocampal extracellular serotonin levels, and a greater antidepressant-like effect in the tail suspension test in Cx43 knock-down (KD) mice bred on a C57BL/6 background compared to their wild-type littermates. Similarly, in outbred Swiss wild-type mice, the intra-hippocampal injection of a shRNA-Cx43 or the acute systemic injection of the Cxs inhibitor carbenoxolone (CBX: 10 mg/kg; i.p.) potentiated the antidepressant-like effects of fluoxetine. Evaluating the effects of such strategies on diazepam (0.5 mg/kg; i.p.), the results indicate that Cx43 KD mice or wild-types injected with a shRNA-Cx43 in the amygdala, but not in the hippocampus, attenuated the anxiolytic-like effects of this benzodiazepine in the elevated plus maze. The chronic systemic administration of CBX mimicked the latter observations. CONCLUSION: Collectively, these data pave the way to the development of potentiating strategies in the field of psychiatry based on the modulation of astroglial Cx43.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Astrocytes/drug effects , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Animals , Astrocytes/metabolism , Benzodiazepines/pharmacology , Diazepam/pharmacology , Fluoxetine/pharmacology , Male , Mice , Mice, Inbred C57BL , Selective Serotonin Reuptake Inhibitors/pharmacology
8.
Epigenetics Chromatin ; 12(1): 63, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601272

ABSTRACT

BACKGROUND: MeCP2-a chromatin-binding protein associated with Rett syndrome-has two main isoforms, MeCP2-E1 and MeCP2-E2, differing in a few N-terminal amino acid residues. Previous studies have shown brain region-specific expression of these isoforms which, in addition to their different cellular localization and differential expression during brain development, suggest that they may also have non-overlapping molecular mechanisms. However, differential functions of MeCP2-E1 and E2 remain largely unexplored. RESULTS: Here, we show that the N-terminal domains (NTD) of MeCP2-E1 and E2 modulate the ability of the methyl-binding domain (MBD) to interact with DNA as well as influencing the turn-over rates, binding dynamics, response to neuronal depolarization, and circadian oscillations of the two isoforms. Our proteomics data indicate that both isoforms exhibit unique interacting protein partners. Moreover, genome-wide analysis using ChIP-seq provide evidence for a shared as well as a specific regulation of different sets of genes. CONCLUSIONS: Our study supports the idea that Rett syndrome might arise from simultaneous impairment of cellular processes involving non-overlapping functions of MECP2 isoforms. For instance, MeCP2-E1 mutations might impact stimuli-dependent chromatin regulation, while MeCP2-E2 mutations could result in aberrant ribosomal expression. Overall, our findings provide insight into the functional complexity of MeCP2 by dissecting differential aspects of its two isoforms.


Subject(s)
DNA/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Circadian Rhythm/genetics , Humans , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Neurons/metabolism , Protein Binding , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Rett Syndrome/genetics , Rett Syndrome/pathology
9.
Proc Natl Acad Sci U S A ; 115(7): 1611-1616, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29378968

ABSTRACT

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.


Subject(s)
Borna Disease/virology , Borna disease virus/physiology , Cognition Disorders/etiology , Hippocampus/virology , Memory, Long-Term/physiology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Viral Structural Proteins/metabolism , Animals , Borna Disease/metabolism , Borna Disease/pathology , Cells, Cultured , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Dentate Gyrus/virology , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mutation , Neuronal Plasticity , Neurons/metabolism , Neurons/pathology , Neurons/virology , Phosphoproteins/genetics , Phosphorylation , Protein Kinase C/genetics , Viral Structural Proteins/genetics
10.
J Gen Virol ; 97(12): 3215-3224, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27902378

ABSTRACT

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.


Subject(s)
Borna Disease/virology , Borna disease virus/metabolism , Endosomes/virology , Neurons/virology , Animals , Borna Disease/metabolism , Borna disease virus/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Endosomes/metabolism , Neurons/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Rats , Rats, Sprague-Dawley , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/metabolism
12.
Biomol Concepts ; 7(2): 103-16, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27101554

ABSTRACT

Cognitive functions require the expression of an appropriate pattern of genes in response to environmental stimuli. Over the last years, many studies have accumulated knowledge towards the understanding of molecular mechanisms that regulate neuronal gene expression. Epigenetic modifications have been shown to play an important role in numerous neuronal functions, from synaptic plasticity to learning and memory. In particular, histone acetylation is a central player in these processes. In this review, we present the molecular mechanisms of histone acetylation and summarize the data underlying the relevance of histone acetylation in cognitive functions in normal and pathological conditions. In the last part, we discuss the different mechanisms underlying the dysregulation of histone acetylation associated with neurological disorders, with a particular focus on environmental causes (stress, drugs, or infectious agents) that are linked to impaired histone acetylation.


Subject(s)
Neurons/physiology , Acetylation , Animals , Cognition , Environment , Epigenesis, Genetic , Gene Expression Regulation , Genetic Predisposition to Disease , Histones/metabolism , Humans , Memory , Mutation , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neuronal Plasticity
13.
J Virol ; 89(11): 5996-6008, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25810554

ABSTRACT

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.


Subject(s)
Borna disease virus/physiology , Epigenesis, Genetic , Host-Pathogen Interactions , Neurons/virology , Phosphoproteins/metabolism , Viral Structural Proteins/metabolism , Virus Replication , Acetylation , Animals , Cells, Cultured , Histones/metabolism , Protein Processing, Post-Translational , Rats, Sprague-Dawley
14.
Virologie (Montrouge) ; 18(4): 187-200, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-33065931

ABSTRACT

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.

15.
J Virol ; 87(22): 12339-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24027309

ABSTRACT

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.


Subject(s)
Borna Disease/virology , Borna disease virus/pathogenicity , Cell Nucleus/metabolism , Cysteine/chemistry , Cytoplasm/metabolism , Phosphoproteins/metabolism , Viral Fusion Proteins/metabolism , Viral Structural Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Borna Disease/metabolism , Chlorocebus aethiops , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Phosphoproteins/genetics , Protein Transport , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Viral Fusion Proteins/genetics , Viral Structural Proteins/genetics
16.
J Biol Chem ; 285(9): 6552-62, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20053986

ABSTRACT

The c-Fos proto-oncogenic transcription factor defines a multigene family controlling many processes both at the cell and the whole organism level. To bind to its target AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element DNA sequences in gene promoters and exert its transcriptional part, c-Fos must heterodimerize with other bZip proteins, its best studied partners being the Jun proteins (c-Jun, JunB, and JunD). c-Fos expression is regulated at many transcriptional and post-transcriptional levels, yet little is known on how its localization is dynamically regulated in the cell. Here we have investigated its intranuclear mobility using fluorescence recovery after photobleaching, genetic, and biochemical approaches. Whereas monomeric c-Fos is highly mobile and distributed evenly with nucleolar exclusion in the nucleus, heterodimerization with c-Jun entails intranuclear redistribution and dramatic reduction in mobility of c-Fos caused by predominant association with the nuclear matrix independently of any binding to AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element sequences. In contrast to c-Jun, dimerization with JunB does not detectably affect c-Fos mobility. However, dimerization with JunB affects intranuclear distribution with significant differences in the localization of c-Fos.c-Jun and c-Fos.JunB dimers. Moreover, c-Jun and JunB exert comparable effects on another Fos family member, Fra-1. Thus, we report a novel regulation, i.e. differentially regulated intranuclear mobility and distribution of Fos proteins by their Jun partners, and suggest the existence of intranuclear storage sites for latent c-Fos.c-Jun AP-1 complexes. This may affect the numerous physiopathological functions these transcription factors control.


Subject(s)
Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/metabolism , HeLa Cells , Humans , Mice , Multiprotein Complexes/metabolism , Protein Multimerization , Protein Transport , Rats , Transcription Factors/metabolism
17.
Stem Cells ; 26(7): 1796-807, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18499894

ABSTRACT

Neural stem cells remain in two areas of the adult mammalian brain, the subventricular zone (SVZ) and the dentate gyrus of the hippocampus. Ongoing neurogenesis via the SVZ-rostral migratory stream pathway maintains neuronal replacement in the olfactory bulb (OB) throughout life. The mechanisms determining how neurogenesis is restricted to only a few regions in the adult, in contrast to its more widespread location during embryogenesis, largely depend on controlling the balance between precursor cell proliferation and differentiation. BM88/Cend1 is a neuronal lineage-specific regulator implicated in cell cycle exit and differentiation of precursor cells in the embryonic neural tube. Here we investigated its role in postnatal neurogenesis. Study of in vivo BM88/Cend1 distribution revealed that it is expressed in low levels in neuronal precursors of the adult SVZ and in high levels in postmitotic OB interneurons. To assess the functional significance of BM88/Cend1 in neuronal lineage progression postnatally, we challenged its expression levels by gain- and loss-of-function approaches using lentiviral gene transfer in SVZ-derived neurospheres. We found that BM88/Cend1 overexpression decreases proliferation and favors neuronal differentiation, whereas its downregulation using new-generation RNA interference vectors yields an opposite phenotype. Our results demonstrate that BM88/Cend1 participates in cell cycle control and neuronal differentiation mechanisms during neonatal SVZ neurogenesis and becomes crucial for the transition from neuroblasts to mature neurons when reaching high levels.


Subject(s)
Hippocampus/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Lineage , Cell Proliferation , Gene Transfer Techniques , Genetic Vectors , Humans , Lentivirus/genetics , Mice , Models, Biological , Swine
18.
J Biol Chem ; 282(42): 31046-59, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17681951

ABSTRACT

c-Fos proto-oncoprotein forms AP-1 transcription complexes with heterodimerization partners such as c-Jun, JunB, and JunD. Thereby, it controls essential cell functions and exerts tumorigenic actions. The dynamics of c-Fos intracellular distribution is poorly understood. Hence, we have combined genetic, cell biology, and microscopic approaches to investigate this issue. In addition to a previously characterized basic nuclear localization signal (NLS) located within the central DNA-binding domain, we identified a second NLS within the c-Fos N-terminal region. This NLS is non-classic and its activity depends on transportin 1 in vivo. Under conditions of prominent nuclear localization, c-Fos can undergo nucleocytoplasmic shuttling through an active Crm-1 exportin-independent mechanism. Dimerization with the Jun proteins inhibits c-Fos nuclear exit. The strongest effect is observed with c-Jun probably in accordance with the relative stabilities of the different c-Fos:Jun dimers. Retrotransport inhibition is not caused by binding of dimers to DNA and, therefore, is not induced by indirect effects linked to activation of c-Fos target genes. Monomeric, but not dimeric, Jun proteins also shuttle actively. Thus, our work unveils a novel regulation operating on AP-1 by demonstrating that dimerization is crucial, not only for active transcription complex formation, but also for keeping them in the compartment where they exert their transcriptional function.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , BALB 3T3 Cells , Cell Nucleus/genetics , Cytoplasm/genetics , Dimerization , HeLa Cells , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factor AP-1/genetics , beta Karyopherins/genetics , beta Karyopherins/metabolism , Exportin 1 Protein
19.
Mol Cell Biol ; 25(16): 6964-79, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16055710

ABSTRACT

The inducible transcriptional complex AP-1, composed of c-Fos and c-Jun proteins, is crucial for cell adaptation to many environmental changes. While its mechanisms of activation have been extensively studied, how its activity is restrained is poorly understood. We report here that lysine 265 of c-Fos is conjugated by the peptidic posttranslational modifiers SUMO-1, SUMO-2, and SUMO-3 and that c-Jun can be sumoylated on lysine 257 as well as on the previously described lysine 229. Sumoylation of c-Fos preferentially occurs in the context of c-Jun/c-Fos heterodimers. Using nonsumoylatable mutants of c-Fos and c-Jun as well as a chimeric protein mimicking sumoylated c-Fos, we show that sumoylation entails lower AP-1 transactivation activity. Interestingly, single sumoylation at any of the three acceptor sites of the c-Fos/c-Jun dimer is sufficient to substantially reduce transcription activation. The lower activity of sumoylated c-Fos is not due to inhibition of protein entry into the nucleus, accelerated turnover, and intrinsic inability to dimerize or to bind to DNA. Instead, cell fractionation experiments suggest that decreased transcriptional activity of sumoylated c-Fos is associated with specific intranuclear distribution. Interestingly, the phosphorylation of threonine 232 observed upon expression of oncogenically activated Ha-Ras is known to superactivate c-Fos transcriptional activity. We show here that it also inhibits c-Fos sumoylation, revealing a functional antagonism between two posttranslational modifications, each occurring within a different moiety of a bipartite transactivation domain of c-Fos. Finally we report that the sumoylation of c-Fos is a dynamic process that can be reversed via multiple mechanisms. This supports the idea that this modification does not constitute a final inactivation step that necessarily precedes protein degradation.


Subject(s)
Down-Regulation , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Binding Sites , Dimerization , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Kinetics , Luciferases/metabolism , Lysine/chemistry , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/chemistry , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Subcellular Fractions , Threonine/chemistry , Time Factors , Transcription Factor AP-1/chemistry , Transcription, Genetic , Transcriptional Activation , Transfection
20.
J Biol Chem ; 279(11): 10261-9, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14672927

ABSTRACT

Initiation of poliovirus (PV) protein synthesis is governed by an internal ribosome entry segment structured into several domains including domain V, which is accepted to be important in PV neurovirulence because it harbors an attenuating mutation in each of the vaccine strains developed by A. Sabin. To better understand how these single point mutations exert their effects, we placed each of them into the same genomic context, that of PV type 1. Only the mutation equivalent to the Sabin type 3 strain mutation resulted in significantly reduced viral growth both in HeLa and neuroblastoma cells. This correlated with poor translation efficiency in vitro and could be explained by a structural perturbation of the domain V of the internal ribosome entry segment, as evidenced by RNA melting experiments. We demonstrated that reduced cell death observed during infection by this mutant is due to the absence of inhibition of host cell translation. We confirmed that this shut-off is correlated principally with cleavage of eIF4GII and not eIF4GI and that this cleavage is significantly impaired in the case of the defective mutant. These data support the previously reported conclusion that the 2A protease has markedly different affinities for the two eIF4G isoforms.


Subject(s)
Mutation , Poliovirus Vaccines/genetics , Poliovirus/genetics , Ribosomes/genetics , Base Sequence , Blotting, Western , DNA, Viral , Electrophoresis, Polyacrylamide Gel , Eukaryotic Initiation Factor-4G/chemistry , HeLa Cells , Humans , Molecular Sequence Data , Neuroblastoma/metabolism , Nucleic Acid Conformation , Phenotype , Plasmids/metabolism , Point Mutation , Protein Biosynthesis , Protein Isoforms , Protein Structure, Tertiary , RNA/chemistry , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...