25 research outputs found

    Etudes structurales sur l'assemblage du nucléosome

    Get PDF
    Assembly of chromatin is an essential process that concerns most DNA transactions in eukaryotic cells. The basic repeating unit of chromatin are nucleosomes, macromolecular complexes that consist of a histone octamer that organizes 147 bp of DNA in two superhelical turns. Although, the structures of nucleosomes are known in detail, their assembly is poorly understood. In vivo, nucleosome assembly is orchestrated by ATP-dependent remodelling enzymes, histone-modifying enzymes and a number of at least partially redundant histone chaperones. Histone chaperons are a structurally diverse class of proteins that direct the productive assembly and disassembly of nucleosomes by facilitating histone deposition and exchange. The currently accepted model is that nucleosome assembly is a sequential process that begins with the interaction of H3/H4 with DNA to form a (H3/H4)2 tetramer-DNA complex. The addition of two H2A/H2B dimers completes a canonical nucleosome. High-resolution structures of histone chaperons in complex with H3/H4 histones have resulted in detailed insights into the process of nucleosome assembly. However, our understanding of the mechanism of nucleosome assembly has been hampered by the as yet limited number of co-crystal structures of histone–chaperone complexes. In particular it remains unclear how histone chaperons mediate H2A/H2B deposition to complete nucleosome assembly. In this work, we have investigated the role of the H2A/H2B chaperon Nap1 (Nucleosome assembly protein 1) in nucleosome assembly. We have determined the crystal structure of the complex between Nap1 and H2A/H2B and analysed the assembly by various biophysical methods. The structure shows that a Nap1 dimer binds to one copy of H2A/H2B (Nap1_2-H2A/H2B). A large ~550 kDa macromolecular assembly containing 6 copies of the Nap12-H2A/H2B complex is seen in the asymmetric crystallographic unit. We confirmed by both non-denaturing mass spectroscopy and negative stain electron microscopy studies that this assembly is the predominant form of the Nap1_2-H2A/H2B complex in solution. We further investigated the potential interplay between p300-mediated histone acetylation and nucleosome assembly. Together, the structure and associated functional analysis provide a detailed mechanism for the Nap1 chaperon activity, its role in H2A/H2B deposition and in nucleosome assembly.Au sein du noyau, l'ADN est organise en chromatine dont l'unité de base est le nucléosome. La structure de la chromatine est très dynamique, ce qui est nécessaire pour la plupart des opérations qui se produisent dans l'ADN telles que la réplication, la transcription, la réparation et la recombinaison. Le nucléosome est constitué de deux dimères H2A/H2B et deux dimères H3/H4 associés avec 147 paires de bases d'ADN. La protéine Nap1 est un chaperon d'histone H2A/H2B impliquée dans l'assemblage et démontage des nucléosomes. Nap1 protège les interactions non spécifiques entre l'ADN chargé négativement et les dimères H2A/H2B chargés positivement, afin de permettre la formation de la structure ordonnée des nucléosomes. Lors de l'assemblage des nucléosomes, les dimères d'histones H3/H4 sont déposés en premier lieu, suivi par le dépôt de dimères H2A/H2B. Lors du démontage du nucléosome, les dimères H2A/H2B sont retirés avant le retrait des dimères H3/H4. La determination de la structure du complexe Nap1-H2A/H2B pourra permettre une meilleure compréhension du processus d'assemblage du nucléosome. Dans cette étude, nous voulons comprendre comment le chaperon Nap1 cible spécifiquement les dimères d'histones H2A/H2B pour l'assemblage des nucléosomes. Notre objectif est de caractériser la structure et la fonction du complexe de Nap1-H2A/H2B. Ainsi nous nous sommes tout d'abord intéresse à la stoechiometrie de ce complexe. Nous avons trouvé qu'un dimère de Nap1 s'associe à un dimère H2A/H2B (Nap1_2-H2A/H2B). D'autre part, l'analyse par spectrométrie de masse non-dénaturante a montré que ce complexe de base peut s'oligomériser et contenir jusqu'à 6 copies de Nap1_2-H2A/H2B. L'analyse de ce complexe par spectrométrie de masse non-dénaturant a montré que ce complexe peu oligomériser dans un grand complexe contenant jusqu'à 6 copies de Nap1_2-H2A/H2B. Nous avons également obtenu la première structure cristalline à basse résolution de ce complexe. L'analyse du même complexe par microscopie électronique à coloration négative a révélé la présence en solution du même oligomère que dans l'unité asymétrique du cristal, qui contient aussi 6 copies de Nap1_2-H2A/H2B. Ainsi, nous avons pu mettre en évidence de nouvelles interfaces d'interaction entre les différents composants de ce complexe qui nous permettent de mieux comprendre le processus d'assemblage des nucléosomes. Le remodelage de la chromatine permet l'expression des gènes eucaryotes. Ce remodelage nécessite des enzymes telles que des histone acétyltransférases (HAT) et les chaperons d'histones. Les HATs acétylent les chaînes latérales des lysines. Il a été proposé que les HATs et les histones chaperons agissent en synergie pour moduler la structure de la chromatine pendant la transcription. La HAT p300 a été proposé d'interagir avec l'histone chaperon Nap1. Nous avons entrepris de caractériser cette interaction. Malheureusement, nos expériences n'ont pas pu détecter d'interaction directe entre ces protéines

    Etudes structurales sur l'assemblage du nucléosome

    Get PDF
    Assembly of chromatin is an essential process that concerns most DNA transactions in eukaryotic cells. The basic repeating unit of chromatin are nucleosomes, macromolecular complexes that consist of a histone octamer that organizes 147 bp of DNA in two superhelical turns. Although, the structures of nucleosomes are known in detail, their assembly is poorly understood. In vivo, nucleosome assembly is orchestrated by ATP-dependent remodelling enzymes, histone-modifying enzymes and a number of at least partially redundant histone chaperones. Histone chaperons are a structurally diverse class of proteins that direct the productive assembly and disassembly of nucleosomes by facilitating histone deposition and exchange. The currently accepted model is that nucleosome assembly is a sequential process that begins with the interaction of H3/H4 with DNA to form a (H3/H4)2 tetramer-DNA complex. The addition of two H2A/H2B dimers completes a canonical nucleosome. High-resolution structures of histone chaperons in complex with H3/H4 histones have resulted in detailed insights into the process of nucleosome assembly. However, our understanding of the mechanism of nucleosome assembly has been hampered by the as yet limited number of co-crystal structures of histone–chaperone complexes. In particular it remains unclear how histone chaperons mediate H2A/H2B deposition to complete nucleosome assembly. In this work, we have investigated the role of the H2A/H2B chaperon Nap1 (Nucleosome assembly protein 1) in nucleosome assembly. We have determined the crystal structure of the complex between Nap1 and H2A/H2B and analysed the assembly by various biophysical methods. The structure shows that a Nap1 dimer binds to one copy of H2A/H2B (Nap1_2-H2A/H2B). A large ~550 kDa macromolecular assembly containing 6 copies of the Nap12-H2A/H2B complex is seen in the asymmetric crystallographic unit. We confirmed by both non-denaturing mass spectroscopy and negative stain electron microscopy studies that this assembly is the predominant form of the Nap1_2-H2A/H2B complex in solution. We further investigated the potential interplay between p300-mediated histone acetylation and nucleosome assembly. Together, the structure and associated functional analysis provide a detailed mechanism for the Nap1 chaperon activity, its role in H2A/H2B deposition and in nucleosome assembly.Au sein du noyau, l'ADN est organise en chromatine dont l'unité de base est le nucléosome. La structure de la chromatine est très dynamique, ce qui est nécessaire pour la plupart des opérations qui se produisent dans l'ADN telles que la réplication, la transcription, la réparation et la recombinaison. Le nucléosome est constitué de deux dimères H2A/H2B et deux dimères H3/H4 associés avec 147 paires de bases d'ADN. La protéine Nap1 est un chaperon d'histone H2A/H2B impliquée dans l'assemblage et démontage des nucléosomes. Nap1 protège les interactions non spécifiques entre l'ADN chargé négativement et les dimères H2A/H2B chargés positivement, afin de permettre la formation de la structure ordonnée des nucléosomes. Lors de l'assemblage des nucléosomes, les dimères d'histones H3/H4 sont déposés en premier lieu, suivi par le dépôt de dimères H2A/H2B. Lors du démontage du nucléosome, les dimères H2A/H2B sont retirés avant le retrait des dimères H3/H4. La determination de la structure du complexe Nap1-H2A/H2B pourra permettre une meilleure compréhension du processus d'assemblage du nucléosome. Dans cette étude, nous voulons comprendre comment le chaperon Nap1 cible spécifiquement les dimères d'histones H2A/H2B pour l'assemblage des nucléosomes. Notre objectif est de caractériser la structure et la fonction du complexe de Nap1-H2A/H2B. Ainsi nous nous sommes tout d'abord intéresse à la stoechiometrie de ce complexe. Nous avons trouvé qu'un dimère de Nap1 s'associe à un dimère H2A/H2B (Nap1_2-H2A/H2B). D'autre part, l'analyse par spectrométrie de masse non-dénaturante a montré que ce complexe de base peut s'oligomériser et contenir jusqu'à 6 copies de Nap1_2-H2A/H2B. L'analyse de ce complexe par spectrométrie de masse non-dénaturant a montré que ce complexe peu oligomériser dans un grand complexe contenant jusqu'à 6 copies de Nap1_2-H2A/H2B. Nous avons également obtenu la première structure cristalline à basse résolution de ce complexe. L'analyse du même complexe par microscopie électronique à coloration négative a révélé la présence en solution du même oligomère que dans l'unité asymétrique du cristal, qui contient aussi 6 copies de Nap1_2-H2A/H2B. Ainsi, nous avons pu mettre en évidence de nouvelles interfaces d'interaction entre les différents composants de ce complexe qui nous permettent de mieux comprendre le processus d'assemblage des nucléosomes. Le remodelage de la chromatine permet l'expression des gènes eucaryotes. Ce remodelage nécessite des enzymes telles que des histone acétyltransférases (HAT) et les chaperons d'histones. Les HATs acétylent les chaînes latérales des lysines. Il a été proposé que les HATs et les histones chaperons agissent en synergie pour moduler la structure de la chromatine pendant la transcription. La HAT p300 a été proposé d'interagir avec l'histone chaperon Nap1. Nous avons entrepris de caractériser cette interaction. Malheureusement, nos expériences n'ont pas pu détecter d'interaction directe entre ces protéines

    Exploring FeLV-Gag-Based VLPs as a New Vaccine Platform-Analysis of Production and Immunogenicity

    Get PDF
    Feline leukemia virus (FeLV) is one of the most prevalent infectious diseases in domestic cats. Although different commercial vaccines are available, none of them provides full protection. Thus, efforts to design a more efficient vaccine are needed. Our group has successfully engineered HIV-1 Gag-based VLPs that induce a potent and functional immune response against the HIV-1 transmembrane protein gp41. Here, we propose to use this concept to generate FeLV-Gag-based VLPs as a novel vaccine strategy against this retrovirus. By analogy to our HIV-1 platform, a fragment of the FeLV transmembrane p15E protein was exposed on FeLV-Gag-based VLPs. After optimization of Gag sequences, the immunogenicity of the selected candidates was evaluated in C57BL/6 and BALB/c mice, showing strong cellular and humoral responses to Gag but failing to generate anti-p15E antibodies. Altogether, this study not only tests the versatility of the enveloped VLP-based vaccine platform but also sheds light on FeLV vaccine research

    A systematic screen for protein–lipid interactions in Saccharomyces cerevisiae

    Get PDF
    Lipids are important cellular metabolites, with a wide range of structural and functional diversity. Many operate as signaling molecules. Lipids though have rarely been studied in large-scale interaction screen; they are poorly represented in current biological networks.Here, we describe the use of miniaturized lipid–arrays for the large-scale study of protein–lipid interactions. In yeast, we show general feasibility with a systematic screen implying 172 proteins. We report 530 protein–lipid associations, the majority is novel and several were validated using other techniques.The screen uncovers numerous insights into lipid function in yeast and equivalent systems in humans. It revealed (i) previously undetected cryptic lipid-binding domains, (ii) series of new cellular targets for sphingolipids and (iii) new ligands for some PH domains that can cooperatively bind additional lipids and work as coincidence sensor to integrate both phosphatidylinositol phosphates and sphingolipid signaling pathways.The significant number of biological insights uncovered shows that even major classes of metabolites have been insufficiently studied. This illustrates the general relevance of such systematic screens and calls for further system-wide analyses

    An engineered HIV-1 Gag-based VLP displaying high antigen density induces strong antibody-dependent functional immune responses

    Get PDF
    Antigen display on the surface of Virus-Like Particles (VLPs) improves immunogenicity compared to soluble proteins. We hypothesised that immune responses can be further improved by increasing the antigen density on the surface of VLPs. In this work, we report an HIV-1 Gag-based VLP platform engineered to maximise the presence of antigen on the VLP surface. An HIV-1 gp41-derived protein (Min), including the C-terminal part of gp41 and the transmembrane domain, was fused to HIV-1 Gag. This resulted in high-density MinGag-VLPs. These VLPs demonstrated to be highly immunogenic in animal models using either a homologous (VLP) or heterologous (DNA/VLP) vaccination regimen, with the latter yielding 10-fold higher anti-Gag and anti-Min antibody titres. Despite these strong humoral responses, immunisation with MinGag-VLPs did not induce neutralising antibodies. Nevertheless, antibodies were predominantly of an IgG2b/IgG2c profile and could efficiently bind CD16-2. Furthermore, we demonstrated that MinGag-VLP vaccination could mediate a functional effect and halt the progression of a Min-expressing tumour cell line in an in vivo mouse model

    SARS-CoV-2 infection elicits a rapid neutralizing antibody response that correlates with disease severity

    Get PDF
    The protective effect of neutralizing antibodies in SARS-CoV-2 infected individuals is not yet well defined. To address this issue, we have analyzed the kinetics of neutralizing antibody responses and their association with disease severity. Between March and May 2020, the prospective KING study enrolled 72 COVID-19+ participants grouped according to disease severity. SARS-CoV-2 infection was diagnosed by serological and virological tests. Plasma neutralizing responses were assessed against replicative virus and pseudoviral particles. Multiple regression and non-parametric tests were used to analyze dependence of parameters. The magnitude of neutralizing titers significantly increased with disease severity. Hospitalized individuals developed higher titers compared to mild-symptomatic and asymptomatic individuals, which together showed titers below the detection limit in 50% of cases. Longitudinal analysis confirmed the strong differences in neutralizing titers between non-hospitalized and hospitalized participants and showed rapid kinetics of appearance of neutralizing antibodies (50% and 80% of maximal activity reached after 11 and 17 days after symptoms onset, respectively) in hospitalized patients. No significant impact of age, gender or treatment on the neutralizing titers was observed in this limited cohort. These data identify a clear association of humoral immunity with disease severity and point to immune mechanisms other than antibodies as relevant players in COVID-19 protection.info:eu-repo/semantics/publishedVersio

    SARS-CoV-2 infection elicits a rapid neutralizing antibody response that correlates with disease severity

    Get PDF
    The protective effect of neutralizing antibodies in SARS-CoV-2 infected individuals is not yet well defined. To address this issue, we have analyzed the kinetics of neutralizing antibody responses and their association with disease severity. Between March and May 2020, the prospective KING study enrolled 72 COVID-19+ participants grouped according to disease severity. SARS-CoV-2 infection was diagnosed by serological and virological tests. Plasma neutralizing responses were assessed against replicative virus and pseudoviral particles. Multiple regression and non-parametric tests were used to analyze dependence of parameters. The magnitude of neutralizing titers significantly increased with disease severity. Hospitalized individuals developed higher titers compared to mild-symptomatic and asymptomatic individuals, which together showed titers below the detection limit in 50% of cases. Longitudinal analysis confirmed the strong differences in neutralizing titers between non-hospitalized and hospitalized participants and showed rapid kinetics of appearance of neutralizing antibodies (50% and 80% of maximal activity reached after 11 and 17 days after symptoms onset, respectively) in hospitalized patients. No significant impact of age, gender or treatment on the neutralizing titers was observed in this limited cohort. These data identify a clear association of humoral immunity with disease severity and point to immune mechanisms other than antibodies as relevant players in COVID-19 protection

    Novel Spike-stabilized trimers with improved production protect K18-hACE2 mice and golden Syrian hamsters from the highly pathogenic SARS-CoV-2 Beta variant

    Get PDF
    Most COVID-19 vaccines are based on the SARS-CoV-2 Spike glycoprotein (S) or their subunits. However, S shows some structural instability that limits its immunogenicity and production, hampering the development of recombinant S-based vaccines. The introduction of the K986P and V987P (S-2P) mutations increases the production and immunogenicity of the recombinant S trimer, suggesting that these two parameters are related. Nevertheless, S-2P still shows some molecular instability and it is produced with low yield. Here we described a novel set of mutations identified by molecular modeling and located in the S2 region of the S-2P that increase its production up to five-fold. Besides their immunogenicity, the efficacy of two representative S-2P-based mutants, S-29 and S-21, protecting from a heterologous SARS-CoV-2 Beta variant challenge was assayed in K18-hACE2 mice (an animal model of severe SARS-CoV-2 disease) and golden Syrian hamsters (GSH) (a moderate disease model). S-21 induced higher level of WH1 and Delta variants neutralizing antibodies than S-2P in K18-hACE2 mice three days after challenge. Viral load in nasal turbinate and oropharyngeal samples were reduced in S-21 and S-29 vaccinated mice. Despite that, only the S-29 protein protected 100% of K18-hACE2 mice from severe disease. When GSH were analyzed, all immunized animals were protected from disease development irrespectively of the immunogen they received. Therefore, the higher yield of S-29, as well as its improved immunogenicity and efficacy protecting from the highly pathogenic SARS-CoV-2 Beta variant, pinpoint the S-29 mutant as an alternative to the S-2P protein for future SARS-CoV-2 vaccine development

    Structural studies of Nucleosome Assembly

    No full text
    Au sein du noyau, l'ADN est organise en chromatine dont l'unité de base est le nucléosome. La structure de la chromatine est très dynamique, ce qui est nécessaire pour la plupart des opérations qui se produisent dans l'ADN telles que la réplication, la transcription, la réparation et la recombinaison. Le nucléosome est constitué de deux dimères H2A/H2B et deux dimères H3/H4 associés avec 147 paires de bases d'ADN. La protéine Nap1 est un chaperon d'histone H2A/H2B impliquée dans l'assemblage et démontage des nucléosomes. Nap1 protège les interactions non spécifiques entre l'ADN chargé négativement et les dimères H2A/H2B chargés positivement, afin de permettre la formation de la structure ordonnée des nucléosomes. Lors de l'assemblage des nucléosomes, les dimères d'histones H3/H4 sont déposés en premier lieu, suivi par le dépôt de dimères H2A/H2B. Lors du démontage du nucléosome, les dimères H2A/H2B sont retirés avant le retrait des dimères H3/H4. La determination de la structure du complexe Nap1-H2A/H2B pourra permettre une meilleure compréhension du processus d'assemblage du nucléosome. Dans cette étude, nous voulons comprendre comment le chaperon Nap1 cible spécifiquement les dimères d'histones H2A/H2B pour l'assemblage des nucléosomes. Notre objectif est de caractériser la structure et la fonction du complexe de Nap1-H2A/H2B. Ainsi nous nous sommes tout d'abord intéresse à la stoechiometrie de ce complexe. Nous avons trouvé qu'un dimère de Nap1 s'associe à un dimère H2A/H2B (Nap1_2-H2A/H2B). D'autre part, l'analyse par spectrométrie de masse non-dénaturante a montré que ce complexe de base peut s'oligomériser et contenir jusqu'à 6 copies de Nap1_2-H2A/H2B. L'analyse de ce complexe par spectrométrie de masse non-dénaturant a montré que ce complexe peu oligomériser dans un grand complexe contenant jusqu'à 6 copies de Nap1_2-H2A/H2B. Nous avons également obtenu la première structure cristalline à basse résolution de ce complexe. L'analyse du même complexe par microscopie électronique à coloration négative a révélé la présence en solution du même oligomère que dans l'unité asymétrique du cristal, qui contient aussi 6 copies de Nap1_2-H2A/H2B. Ainsi, nous avons pu mettre en évidence de nouvelles interfaces d'interaction entre les différents composants de ce complexe qui nous permettent de mieux comprendre le processus d'assemblage des nucléosomes. Le remodelage de la chromatine permet l'expression des gènes eucaryotes. Ce remodelage nécessite des enzymes telles que des histone acétyltransférases (HAT) et les chaperons d'histones. Les HATs acétylent les chaînes latérales des lysines. Il a été proposé que les HATs et les histones chaperons agissent en synergie pour moduler la structure de la chromatine pendant la transcription. La HAT p300 a été proposé d'interagir avec l'histone chaperon Nap1. Nous avons entrepris de caractériser cette interaction. Malheureusement, nos expériences n'ont pas pu détecter d'interaction directe entre ces protéines.Assembly of chromatin is an essential process that concerns most DNA transactions in eukaryotic cells. The basic repeating unit of chromatin are nucleosomes, macromolecular complexes that consist of a histone octamer that organizes 147 bp of DNA in two superhelical turns. Although, the structures of nucleosomes are known in detail, their assembly is poorly understood. In vivo, nucleosome assembly is orchestrated by ATP-dependent remodelling enzymes, histone-modifying enzymes and a number of at least partially redundant histone chaperones. Histone chaperons are a structurally diverse class of proteins that direct the productive assembly and disassembly of nucleosomes by facilitating histone deposition and exchange. The currently accepted model is that nucleosome assembly is a sequential process that begins with the interaction of H3/H4 with DNA to form a (H3/H4)2 tetramer-DNA complex. The addition of two H2A/H2B dimers completes a canonical nucleosome. High-resolution structures of histone chaperons in complex with H3/H4 histones have resulted in detailed insights into the process of nucleosome assembly. However, our understanding of the mechanism of nucleosome assembly has been hampered by the as yet limited number of co-crystal structures of histone–chaperone complexes. In particular it remains unclear how histone chaperons mediate H2A/H2B deposition to complete nucleosome assembly. In this work, we have investigated the role of the H2A/H2B chaperon Nap1 (Nucleosome assembly protein 1) in nucleosome assembly. We have determined the crystal structure of the complex between Nap1 and H2A/H2B and analysed the assembly by various biophysical methods. The structure shows that a Nap1 dimer binds to one copy of H2A/H2B (Nap1_2-H2A/H2B). A large ~550 kDa macromolecular assembly containing 6 copies of the Nap12-H2A/H2B complex is seen in the asymmetric crystallographic unit. We confirmed by both non-denaturing mass spectroscopy and negative stain electron microscopy studies that this assembly is the predominant form of the Nap1_2-H2A/H2B complex in solution. We further investigated the potential interplay between p300-mediated histone acetylation and nucleosome assembly. Together, the structure and associated functional analysis provide a detailed mechanism for the Nap1 chaperon activity, its role in H2A/H2B deposition and in nucleosome assembly

    Etudes structurales sur l'assemblage du nucléosome

    No full text
    Au sein du noyau, l'ADN est organise en chromatine dont l'unité de base est le nucléosome. La structure de la chromatine est très dynamique, ce qui est nécessaire pour la plupart des opérations qui se produisent dans l'ADN telles que la réplication, la transcription, la réparation et la recombinaison. Le nucléosome est constitué de deux dimères H2A/H2B et deux dimères H3/H4 associés avec 147 paires de bases d'ADN. La protéine Nap1 est un chaperon d'histone H2A/H2B impliquée dans l'assemblage et démontage des nucléosomes. Nap1 protège les interactions non spécifiques entre l'ADN chargé négativement et les dimères H2A/H2B chargés positivement, afin de permettre la formation de la structure ordonnée des nucléosomes. Lors de l'assemblage des nucléosomes, les dimères d'histones H3/H4 sont déposés en premier lieu, suivi par le dépôt de dimères H2A/H2B. Lors du démontage du nucléosome, les dimères H2A/H2B sont retirés avant le retrait des dimères H3/H4. La determination de la structure du complexe Nap1-H2A/H2B pourra permettre une meilleure compréhension du processus d'assemblage du nucléosome. Dans cette étude, nous voulons comprendre comment le chaperon Nap1 cible spécifiquement les dimères d'histones H2A/H2B pour l'assemblage des nucléosomes. Notre objectif est de caractériser la structure et la fonction du complexe de Nap1-H2A/H2B. Ainsi nous nous sommes tout d'abord intéresse à la stoechiometrie de ce complexe. Nous avons trouvé qu'un dimère de Nap1 s'associe à un dimère H2A/H2B (Nap1_2-H2A/H2B). D'autre part, l'analyse par spectrométrie de masse non-dénaturante a montré que ce complexe de base peut s'oligomériser et contenir jusqu'à 6 copies de Nap1_2-H2A/H2B. L'analyse de ce complexe par spectrométrie de masse non-dénaturant a montré que ce complexe peu oligomériser dans un grand complexe contenant jusqu'à 6 copies de Nap1_2-H2A/H2B. Nous avons également obtenu la première structure cristalline à basse résolution de ce complexe. L'analyse du même complexe par microscopie électronique à coloration négative a révélé la présence en solution du même oligomère que dans l'unité asymétrique du cristal, qui contient aussi 6 copies de Nap1_2-H2A/H2B. Ainsi, nous avons pu mettre en évidence de nouvelles interfaces d'interaction entre les différents composants de ce complexe qui nous permettent de mieux comprendre le processus d'assemblage des nucléosomes. Le remodelage de la chromatine permet l'expression des gènes eucaryotes. Ce remodelage nécessite des enzymes telles que des histone acétyltransférases (HAT) et les chaperons d'histones. Les HATs acétylent les chaînes latérales des lysines. Il a été proposé que les HATs et les histones chaperons agissent en synergie pour moduler la structure de la chromatine pendant la transcription. La HAT p300 a été proposé d'interagir avec l'histone chaperon Nap1. Nous avons entrepris de caractériser cette interaction. Malheureusement, nos expériences n'ont pas pu détecter d'interaction directe entre ces protéines.Assembly of chromatin is an essential process that concerns most DNA transactions in eukaryotic cells. The basic repeating unit of chromatin are nucleosomes, macromolecular complexes that consist of a histone octamer that organizes 147 bp of DNA in two superhelical turns. Although, the structures of nucleosomes are known in detail, their assembly is poorly understood. In vivo, nucleosome assembly is orchestrated by ATP-dependent remodelling enzymes, histone-modifying enzymes and a number of at least partially redundant histone chaperones. Histone chaperons are a structurally diverse class of proteins that direct the productive assembly and disassembly of nucleosomes by facilitating histone deposition and exchange. The currently accepted model is that nucleosome assembly is a sequential process that begins with the interaction of H3/H4 with DNA to form a (H3/H4)2 tetramer-DNA complex. The addition of two H2A/H2B dimers completes a canonical nucleosome. High-resolution structures of histone chaperons in complex with H3/H4 histones have resulted in detailed insights into the process of nucleosome assembly. However, our understanding of the mechanism of nucleosome assembly has been hampered by the as yet limited number of co-crystal structures of histone chaperone complexes. In particular it remains unclear how histone chaperons mediate H2A/H2B deposition to complete nucleosome assembly. In this work, we have investigated the role of the H2A/H2B chaperon Nap1 (Nucleosome assembly protein 1) in nucleosome assembly. We have determined the crystal structure of the complex between Nap1 and H2A/H2B and analysed the assembly by various biophysical methods. The structure shows that a Nap1 dimer binds to one copy of H2A/H2B (Nap1_2-H2A/H2B). A large ~550 kDa macromolecular assembly containing 6 copies of the Nap12-H2A/H2B complex is seen in the asymmetric crystallographic unit. We confirmed by both non-denaturing mass spectroscopy and negative stain electron microscopy studies that this assembly is the predominant form of the Nap1_2-H2A/H2B complex in solution. We further investigated the potential interplay between p300-mediated histone acetylation and nucleosome assembly. Together, the structure and associated functional analysis provide a detailed mechanism for the Nap1 chaperon activity, its role in H2A/H2B deposition and in nucleosome assembly.SAVOIE-SCD - Bib.électronique (730659901) / SudocGRENOBLE1/INP-Bib.électronique (384210012) / SudocGRENOBLE2/3-Bib.électronique (384219901) / SudocSudocFranceF
    corecore