Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 32
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Environ Toxicol Pharmacol ; 111: 104563, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39260711

RÉSUMÉ

Micro- and nanoplastics (MPs/NPs) constitute emerging and widely-distributed environmental contaminants to which humans are highly exposed. They possibly represent a threat for human health. In order to identify cellular/molecular targets for these plastic particles, we have analysed the effects of exposure to manufactured polystyrene (PS) MPs and NPs on in vitro activity and expression of human membrane drug transporters, known to interact with chemical pollutants. PS MPs and NPs, used at various concentrations (1, 10 or 100 µg/mL), failed to inhibit efflux activities of the ATP-binding cassette (ABC) transporters P-glycoprotein, MRPs and BCRP in ABC transporter-expressing cells. Furthermore, PS particles did not impair the transport of P-glycoprotein or BCRP substrates across intestinal Caco-2 cell monolayers. Uptake activities of solute carriers (SLCs) such as OCT1 and OCT2 (handling organic cations) or OATP1B1, OATP1B3, OATP2B1, OAT1 and OAT3 (handling organic anions) were additionally not altered by PS MPs/NPs in HEK-293 cells overexpressing these SLCs. mRNA expression of ABC transporters and of the SLCs OCT1 and OATP2B1 in Caco-2 cells and human hepatic HepaRG cells were finally not impaired by a 48-h exposure to MPs/NPs. Altogether, these data indicate that human drug transporters are unlikely to be direct and univocal targets for synthetic PS MPs/NPs.

2.
Exp Parasitol ; 265: 108811, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39111383

RÉSUMÉ

Alveolar echinococcosis (AE) is a severe disease caused by the infection with the larval stage of Echinococcus multilocularis, the metacestode. As there is no actual curative drug therapy, recommendations to manage AE patients are based on radical surgery and prophylactic administration of albendazole or mebendazole during 2 years to prevent relapses. There is an urgent need for new therapeutic strategies for the management of AE, as the drugs in use are only parasitostatic, and can induce toxicity. This study aimed at developing a drug delivery system for mefloquine, an antiparasitic compound which is highly active against E. multilocularis in vitro and in experimentally infected mice. We formulated mefloquine-loaded PLGA-PEG-COOH (poly-(lactic-co-glycolic acid)) nanoparticles that exhibit stable physical properties and mefloquine content. These nanoparticles crossed the outer acellular laminated layer of metacestodes in vitro and delivered their content to the inner germinal layer within less than 5 min. The in vitro anti-echinococcal activity of mefloquine was not altered during the formulation process. However, toxicity against hepatocytes was not reduced when compared to free mefloquine. Altogether, this study shows that mefloquine-loaded PLGA-PEG-COOH nanoparticles are promising candidates for drug delivery during AE treatment. However, strategies for direct parasite-specific targeting of these particles should be developed.


Sujet(s)
Echinococcus multilocularis , Méfloquine , Nanoparticules , Polyéthylène glycols , Animaux , Méfloquine/pharmacologie , Méfloquine/administration et posologie , Echinococcus multilocularis/effets des médicaments et des substances chimiques , Souris , Polyéthylène glycols/composition chimique , Nanoparticules/composition chimique , Échinococcose/traitement médicamenteux , Systèmes de délivrance de médicaments , Femelle , Souris de lignée BALB C , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Anthelminthiques/pharmacologie , Anthelminthiques/administration et posologie , Anthelminthiques/composition chimique , Humains , Polyglactine 910
4.
FEBS Lett ; 598(7): 758-773, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38436147

RÉSUMÉ

The human Mediator complex subunit MED25 binds transactivation domains (TADs) present in various cellular and viral proteins using two binding interfaces, named H1 and H2, which are found on opposite sides of its ACID domain. Here, we use and compare deep learning methods to characterize human MED25-TAD interfaces and assess the predicted models to published experimental data. For the H1 interface, AlphaFold produces predictions with high-reliability scores that agree well with experimental data, while the H2 interface predictions appear inconsistent, preventing reliable binding modes. Despite these limitations, we experimentally assess the validity of MED25 interface predictions with the viral transcriptional activators Lana-1 and IE62. AlphaFold predictions also suggest the existence of a unique hydrophobic pocket for the Arabidopsis MED25 ACID domain.


Sujet(s)
Protéines précoces immédiates , Complexe médiateur , Humains , Complexe médiateur/génétique , Complexe médiateur/métabolisme , Activation de la transcription , Reproductibilité des résultats , Facteurs de transcription/métabolisme , Protéines de l'enveloppe virale/métabolisme , Transactivateurs/métabolisme , Protéines précoces immédiates/métabolisme
5.
Trends Biochem Sci ; 46(9): 705-707, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34103236

RÉSUMÉ

Cryo-electron microscopy has enabled unprecedented progress in the quest to reveal the structure of the whole transcription preinitiation complex. Four recent studies pave the way for a complete description of how transcription is initiated at near-atomic level.


Sujet(s)
Complexe médiateur , RNA polymerase II , Cryomicroscopie électronique , Complexe médiateur/génétique , RNA polymerase II/métabolisme , Transcription génétique
6.
J Biol Chem ; 295(39): 13617-13629, 2020 09 25.
Article de Anglais | MEDLINE | ID: mdl-32737196

RÉSUMÉ

The evolutionarily conserved multiprotein Mediator complex (MED) serves as an interface between DNA-bound transcription factors (TFs) and the RNA Pol II machinery. It has been proposed that each TF interacts with a dedicated MED subunit to induce specific transcriptional responses. But are these binary partnerships sufficient to mediate TF functions? We have previously established that the Med1 Mediator subunit serves as a cofactor of GATA TFs in Drosophila, as shown in mammals. Here, we observe mutant phenotype similarities between another subunit, Med19, and the Drosophila GATA TF Pannier (Pnr), suggesting functional interaction. We further show that Med19 physically interacts with the Drosophila GATA TFs, Pnr and Serpent (Srp), in vivo and in vitro through their conserved C-zinc finger domains. Moreover, Med19 loss of function experiments in vivo or in cellulo indicate that it is required for Pnr- and Srp-dependent gene expression, suggesting general GATA cofactor functions. Interestingly, Med19 but not Med1 is critical for the regulation of all tested GATA target genes, implying shared or differential use of MED subunits by GATAs depending on the target gene. Lastly, we show a direct interaction between Med19 and Med1 by GST pulldown experiments indicating privileged contacts between these two subunits of the MED middle module. Together, these findings identify Med19/Med1 as a composite GATA TF interface and suggest that binary MED subunit-TF partnerships are probably oversimplified models. We propose several mechanisms to account for the transcriptional regulation of GATA-targeted genes.


Sujet(s)
Protéines de Drosophila/métabolisme , Facteurs de transcription GATA/métabolisme , Complexe médiateur/métabolisme , Animaux , Sites de fixation , Protéines de Drosophila/génétique , Drosophila melanogaster , Facteurs de transcription GATA/génétique , Régulation de l'expression des gènes/génétique
7.
Biochem Soc Trans ; 47(1): 399-410, 2019 02 28.
Article de Anglais | MEDLINE | ID: mdl-30733343

RÉSUMÉ

Mediator is a large multiprotein complex conserved in all eukaryotes that plays an essential role in transcriptional regulation. Mediator comprises 25 subunits in yeast and 30 subunits in humans that form three main modules and a separable four-subunit kinase module. For nearly 20 years, because of its size and complexity, Mediator has posed a formidable challenge to structural biologists. The first two-dimensional electron microscopy (EM) projection map of Mediator leading to the canonical view of its division in three topological modules named Head, Middle and Tail, was published in 1999. Within the last few years, optimization of Mediator purification combined with technical and methodological advances in cryo-electron microscopy (cryo-EM) have revealed unprecedented details of Mediator subunit organization, interactions with RNA polymerase II and parts of its core structure at high resolution. To celebrate the twentieth anniversary of the first Mediator EM reconstruction, we look back on the structural studies of Mediator complex from a historical perspective and discuss them in the light of our current understanding of its role in transcriptional regulation.


Sujet(s)
Complexe médiateur/composition chimique , Cryomicroscopie électronique , Humains , Conformation des protéines , Protéines de Saccharomyces cerevisiae/composition chimique , Protéines de Schizosaccharomyces pombe/composition chimique
8.
Nat Commun ; 9(1): 3389, 2018 08 23.
Article de Anglais | MEDLINE | ID: mdl-30140054

RÉSUMÉ

The Mediator complex transduces regulatory information from enhancers to promoters and performs essential roles in the initiation of transcription in eukaryotes. Human Mediator comprises 26 subunits forming three modules termed Head, Middle and Tail. Here we present the 2.8 Å crystal structure of MED23, the largest subunit from the human Tail module. The structure identifies 25 HEAT repeats-like motifs organized into 5 α-solenoids. MED23 adopts an arch-shaped conformation, with an N-terminal domain (Nter) protruding from a large core region. In the core four solenoids, motifs wrap on themselves, creating triangular-shaped structural motifs on both faces of the arch, with extended grooves propagating through the interfaces between the solenoid motifs. MED23 is known to interact with several specific transcription activators and is involved in splicing, elongation, and post-transcriptional events. The structure rationalizes previous biochemical observations and paves the way for improved understanding of the cross-talk between Mediator and transcriptional activators.


Sujet(s)
Complexe médiateur/composition chimique , Sous-unités de protéines/composition chimique , Motifs d'acides aminés , Cristallisation , Cristallographie aux rayons X , Humains , Complexe médiateur/métabolisme , Domaines protéiques , Sous-unités de protéines/métabolisme , Protéines recombinantes/biosynthèse , Anticorps à domaine unique/métabolisme
9.
Int J Nanomedicine ; 12: 8427-8442, 2017.
Article de Anglais | MEDLINE | ID: mdl-29200853

RÉSUMÉ

Decitabine is a hydrophilic drug that acts by hypomethylating DNA. Decitabine is used in Europe for the treatment of acute myeloid leukemia (AML) in patients aged ≥65 years. However, it can only be administered intravenously due to very low oral bioavailability and a large distribution volume. Oral administration would allow outpatient treatment, improving quality of life and reducing treatment costs. The present study proposes to develop lipid nanocapsules (LNCs), originally designed for lipophilic drugs, to encapsulate decitabine. Two different formulations of LNCs were designed: LNCs based on a high proportion of Transcutol® HP (THP-LNCs) and LNCs associated with a mixture of Transcutol® HP and Tween® 80 (THP-T80-LNCs). The second formulation had a diameter of 26.5±0.5 nm, high encapsulation efficiency (>85%), and a drug payload of 472±64 µg/mL. Decitabine-loaded THP-T80-LNC cytotoxicity was evaluated on two AML cell lines depending on their decitabine resistance: HEL (not resistant) and HL-60 (resistant). The permeability of decitabine-loaded THP-T80-LNCs was also evaluated on Caco-2 cell monolayers. Decitabine cytotoxicity against HEL and HL-60 was higher when decitabine was loaded in THP-T80-LNCs than when free. Apparent permeability on Caco-2 cell monolayers was also increased, suggesting a potentially useful formulation to increase the oral bioavailability of decitabine.


Sujet(s)
Azacitidine/analogues et dérivés , Vecteurs de médicaments/composition chimique , Leucémie aigüe myéloïde/traitement médicamenteux , Nanocapsules/administration et posologie , Administration par voie orale , Antimétabolites antinéoplasiques/administration et posologie , Antimétabolites antinéoplasiques/pharmacocinétique , Azacitidine/administration et posologie , Azacitidine/pharmacocinétique , Biodisponibilité , Cellules Caco-2 , Lignée cellulaire tumorale , Décitabine , Vecteurs de médicaments/administration et posologie , Libération de médicament , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Stabilité de médicament , Éthylène glycols/composition chimique , Humains , Lipides/composition chimique , Nanocapsules/composition chimique , Polysorbates/composition chimique
10.
J Mol Biol ; 429(20): 3043-3055, 2017 10 13.
Article de Anglais | MEDLINE | ID: mdl-28893534

RÉSUMÉ

MED26 is a subunit of Mediator, a large complex central to the regulation of gene transcription by RNA Polymerase II. MED26 plays a role in the switch between the initiation and elongation phases of RNA Polymerase II-mediated transcription process. Regulation of these steps requires successive binding of MED26 N-terminal domain (NTD) to TATA-binding protein-associated factor 7 (TAF7) and Eleven-nineteen lysine-rich in leukemia-Associated Factor 1 (EAF1). In order to investigate the mechanism of regulation by MED26, MED26-NTD structure was solved by NMR, revealing a 4-helix bundle. EAF1 (239-268) and TAF7 (205-235) peptide interactions were both mapped to the same groove formed by H3 and H4 helices of MED26-NTD. Both interactions are characterized by dissociation constants in the 10-µM range. Further experiments revealed a folding-upon-binding mechanism that leads to the formation of EAF1 (N247-S260) and TAF7 (L214-S227) helices. Chemical shift perturbations and nuclear Overhauser enhancement contacts support the involvement of residues I222/F223 in anchoring TAF7 helix to a hydrophobic pocket of MED26-NTD, including residues L48, W80 and I84. In addition, Ala mutations of charged residues located in the C-terminal disordered part of TAF7 and EAF1 peptides affected the binding, with a loss of affinity characterized by a 10-time increase of dissociation constants. A structural model of MED26-NTD/TAF7 complex shows bi-partite components, combining ordered and disordered segments, as well as hydrophobic and electrostatic contributions to the binding. This study provides molecular detail that will help to decipher the mechanistic basis for the initiation to elongation switch-function mediated by MED26-NTD.


Sujet(s)
Complexe médiateur/composition chimique , Complexe médiateur/métabolisme , Facteurs associés à la protéine de liaison à la boite TATA/composition chimique , Facteurs associés à la protéine de liaison à la boite TATA/métabolisme , Facteur de transcription TFIID/composition chimique , Facteur de transcription TFIID/métabolisme , Facteurs de transcription/métabolisme , Humains , Spectroscopie par résonance magnétique , Liaison aux protéines , Conformation des protéines , Cartographie d'interactions entre protéines
11.
Biomol NMR Assign ; 10(1): 233-6, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26861138

RÉSUMÉ

MED26 is a subunit of the Mediator, a very large complex involved in regulation of gene transcription by RNA Polymerase II. MED26 regulates the switch between initiation and elongation phases of the transcription. This function requires interaction of its N-terminal domain (NTD) with several protein partners implicated in transcriptional regulation. Molecular details of the structure and interaction mode of MED26 NTD would improve understanding of this complex regulation. As a first step towards structural characterization, sequence specific (1)H, (13)C and (15)N assignments for MED26 NTD was performed based on Nuclear Magnetic Resonance spectroscopy. TALOS+ analysis of the chemical shifts data revealed a domain solely composed of helices. Assignments will be further used to solve NMR structure and dynamics of MED26 NTD and investigate the molecular details of its interaction with protein partners.


Sujet(s)
Complexe médiateur/composition chimique , Résonance magnétique nucléaire biomoléculaire , Sous-unités de protéines/composition chimique , Séquence d'acides aminés , Complexe médiateur/métabolisme , Domaines protéiques , Structure secondaire des protéines , Sous-unités de protéines/métabolisme
13.
J Biol Chem ; 290(38): 23307-19, 2015 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-26203186

RÉSUMÉ

The two-component sensory transduction system BvgAS controls the virulence regulon of the whooping-cough agent Bordetella pertussis. The periplasmic moiety of the homodimeric sensor kinase BvgS is composed of four bilobed Venus flytrap (VFT) perception domains followed by α helices that extend into the cytoplasmic membrane. In the virulent phase, the default state of B. pertussis, the cytoplasmic enzymatic moiety of BvgS acts as kinase by autophosphorylating and transferring the phosphoryl group to the response regulator BvgA. Under laboratory conditions, BvgS shifts to phosphatase activity in response to modulators, notably nicotinate ions. Here we characterized the effects of nicotinate and related modulators on the BvgS periplasmic moiety by using site-directed mutagenesis and in silico and biophysical approaches. Modulators bind with low affinity to BvgS in the VFT2 cavity. Electron paramagnetic resonance shows that their binding globally affects the conformation and dynamics of the periplasmic moiety. Specific amino acid substitutions designed to slacken interactions within and between the VFT lobes prevent BvgS from responding to nicotinate, showing that BvgS shifts from kinase to phosphatase activity in response to this modulator via a tense transition state that involves a large periplasmic structural block. We propose that this transition enables the transmembrane helices to adopt a distinct conformation that sets the cytoplasmic enzymatic moiety in the phosphatase mode. The bona fide, in vivo VFT ligands that remain to be identified are likely to trigger similar effects on the transmembrane and cytoplasmic moieties. This mechanism may be relevant to the other VFT-containing sensor kinases homologous to BvgS.


Sujet(s)
Protéines bactériennes/métabolisme , Bordetella pertussis/enzymologie , Membrane cellulaire/enzymologie , Acide nicotinique/métabolisme , Protein kinases/métabolisme , Transduction du signal/physiologie , Protéines bactériennes/génétique , Bordetella pertussis/génétique , Membrane cellulaire/génétique , Acide nicotinique/génétique , Périplasme/enzymologie , Périplasme/génétique , Phosphoprotein Phosphatases/génétique , Phosphoprotein Phosphatases/métabolisme , Protein kinases/génétique , Structure secondaire des protéines , Structure tertiaire des protéines
14.
Nucleic Acids Res ; 43(14): 7110-21, 2015 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-26130716

RÉSUMÉ

The N-terminal acidic transactivation domain (TAD) of ERM/ETV5 (ERM38-68), a PEA3 group member of Ets-related transcription factors, directly interacts with the ACID/PTOV domain of the Mediator complex subunit MED25. Molecular details of this interaction were investigated using nuclear magnetic resonance (NMR) spectroscopy. The TAD is disordered in solution but has a propensity to adopt local transient secondary structure. We show that it folds upon binding to MED25 and that the resulting ERM-MED25 complex displays characteristics of a fuzzy complex. Mutational analysis further reveals that two aromatic residues in the ERM TAD (F47 and W57) are involved in the binding to MED25 and participate in the ability of ERM TAD to activate transcription. Mutation of a key residue Q451 in the VP16 H1 binding pocket of MED25 affects the binding of ERM. Furthermore, competition experiments show that ERM and VP16 H1 share a common binding interface on MED25. NMR data confirms the occupancy of this binding pocket by ERM TAD. Based on these experimental data, a structural model of a functional interaction is proposed. This study provides mechanistic insights into the Mediator-transactivator interactions.


Sujet(s)
Protéines de liaison à l'ADN/composition chimique , Complexe médiateur/composition chimique , Transactivateurs/composition chimique , Facteurs de transcription/composition chimique , Protéines de liaison à l'ADN/métabolisme , Complexe médiateur/génétique , Complexe médiateur/métabolisme , Modèles moléculaires , Mutation , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Transactivateurs/métabolisme , Facteurs de transcription/métabolisme
15.
Nucleic Acids Res ; 41(9): 4847-59, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23531547

RÉSUMÉ

PEA3, ERM and ER81 belong to the PEA3 subfamily of Ets transcription factors and play important roles in a number of tissue-specific processes. Transcriptional activation by PEA3 subfamily factors requires their characteristic amino-terminal acidic transactivation domain (TAD). However, the cellular targets of this domain remain largely unknown. Using ERM as a prototype, we show that the minimal N-terminal TAD activates transcription by contacting the activator interacting domain (ACID)/Prostate tumor overexpressed protein 1 (PTOV) domain of the Mediator complex subunit MED25. We further show that depletion of MED25 disrupts the association of ERM with the Mediator in vitro. Small interfering RNA-mediated knockdown of MED25 as well as the overexpression of MED25-ACID and MED25-VWA domains efficiently inhibit the transcriptional activity of ERM. Moreover, mutations of amino acid residues that prevent binding of MED25 to ERM strongly reduce transactivation by ERM. Finally we show that siRNA depletion of MED25 diminishes PEA3-driven expression of MMP-1 and Mediator recruitment. In conclusion, this study identifies the PEA3 group members as the first human transcriptional factors that interact with the MED25 ACID/PTOV domain and establishes MED25 as a crucial transducer of their transactivation potential.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Complexe médiateur/métabolisme , Facteurs de transcription/métabolisme , Activation de la transcription , Lignée cellulaire , Protéines de liaison à l'ADN/composition chimique , Humains , Complexe médiateur/composition chimique , Complexe médiateur/génétique , Mutation , Motifs et domaines d'intéraction protéique , Facteurs de transcription/composition chimique
16.
Article de Anglais | MEDLINE | ID: mdl-22949187

RÉSUMÉ

Two-component and phosphorelay signal-transduction proteins are crucial for bacterial cell-cycle regulation in Caulobacter crescentus. ChpT is an essential histidine phosphotransferase that controls the activity of the master cell-cycle regulator CtrA by phosphorylation. Here, the 2.2 Å resolution crystal structure of ChpT is reported. ChpT is a homodimer and adopts the domain architecture of the intracellular part of class I histidine kinases. Each subunit consists of two distinct domains: an N-terminal helical hairpin domain and a C-terminal α/ß domain. The two N-terminal domains are adjacent within the dimer, forming a four-helix bundle. The ChpT C-terminal domain adopts an atypical Bergerat ATP-binding fold.


Sujet(s)
Caulobacter crescentus/enzymologie , Cycle cellulaire , Phosphotransferases/composition chimique , Structure quaternaire des protéines , Séquence d'acides aminés , Caulobacter crescentus/cytologie , Modèles moléculaires , Données de séquences moléculaires , Structure tertiaire des protéines
17.
J Biol Chem ; 287(2): 1306-21, 2012 Jan 06.
Article de Anglais | MEDLINE | ID: mdl-22128182

RÉSUMÉ

Growing evidence indicates that the protein regulators governing protein phosphatase 1 (PP1) activity have crucial functions because their deletion drastically affects cell growth and division. PP1 has been found to be essential in Plasmodium falciparum, but little is known about its regulators. In this study, we have identified a homolog of Inhibitor-3 of PP1, named PfI3. NMR analysis shows that PfI3 belongs to the disordered protein family. High affinity interaction of PfI3 and PfPP1 is demonstrated in vitro using several methods, with an apparent dissociation constant K(D) of 100 nm. We further show that the conserved (41)KVVRW(45) motif is crucial for this interaction as the replacement of the Trp(45) by an Ala(45) severely decreases the binding to PfPP1. Surprisingly, PfI3 was unable to rescue a yeast strain deficient in I3 (Ypi1). This lack of functional orthology was supported as functional assays in vitro have revealed that PfI3, unlike yeast I3 and human I3, increases PfPP1 activity. Reverse genetic approaches suggest an essential role of PfI3 in the growth and/or survival of blood stage parasites because attempts to obtain knock-out parasites were unsuccessful, although the locus of PfI3 is accessible. The main localization of a GFP-tagged PfI3 in the nucleus of all blood stage parasites is compatible with a regulatory role of PfI3 on the activity of nuclear PfPP1.


Sujet(s)
Noyau de la cellule/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Plasmodium falciparum/métabolisme , Protein Phosphatase 1/antagonistes et inhibiteurs , Protein Phosphatase 1/métabolisme , Protéines de protozoaire/métabolisme , Noyau de la cellule/génétique , Antienzymes/métabolisme , Test de complémentation , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Plasmodium falciparum/génétique , Liaison aux protéines , Protein Phosphatase 1/génétique , Protéines de protozoaire/antagonistes et inhibiteurs , Protéines de protozoaire/génétique , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme
18.
Biochem J ; 439(3): 469-77, 2011 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-21736557

RÉSUMÉ

The PEA3 (polyoma enhancer activator 3) group members [ERM (ETS-related molecule), ER81 (ETS-related 81) and PEA3] of the Ets transcription factor family are involved in migration and dissemination processes during organogenesis and cancer development. In the present study, we report that the hnRNP (heterogeneous nuclear ribonucleoprotein)-like protein CoAA (Coactivator activator) interacts with the PEA3 group members and modulates their transcriptional activity. We also demonstrate that the CoAA YQ domain, containing tyrosine/glutamine-rich hexapeptide repeats, is necessary for the interaction, whereas the two N-terminal RRMs (RNA recognition motifs) of CoAA are required to enhance transcriptional activity. Finally, we show that CoAA is involved in the migration-enhancing action of PEA3 on MCF7 human cancer cells, suggesting that CoAA might be an important regulator of PEA3 group member activity during metastasis.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/physiologie , Facteurs de transcription/biosynthèse , Activation de la transcription/physiologie , Animaux , Mouvement cellulaire/génétique , Cellules HEK293 , Cellules HeLa , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Lapins , Facteurs de transcription/génétique
19.
Protein Expr Purif ; 80(2): 211-6, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21708266

RÉSUMÉ

Covalent modification of proteins with SUMO (Small Ubiquitin-like MOdifier) affects many cellular processes, including transcriptional regulation, DNA repair and signal transduction. Although hundreds of SUMO targets have been identified, many biological outcomes of protein sumoylation remain poorly understood. In particular, biochemical and structural analysis can only be easily conducted if highly pure sumoylated substrates are available. Purification of sumoylated substrates in vitro or in bacteria have been previously reported but separating the sumoylated protein from the undesired unmodified fraction is often technically challenging, inefficient and time consuming. Here we develop a new vector system for in vivo sumoylation in Escherichia coli which improves purification of sumoylated proteins. We describe the purification of IκBα, its sumoylation, the subsequent separation and purification of the modified and the unmodified forms and the purification of the complex IκBα-SUMO-1/NF-κB. After a first GST affinity chromatography and GST-tag removal, a unique metal-ion affinity chromatography using a 6xHis-SUMO-1 tag results in mgs of highly pure SUMO-1 modified IκBα. Our pure SUMO-1 modified IκB/NF-κB complex could be a useful tool to identify new interaction partner specific of the SUMO-1 modified IκBα form. This approach may be extended to other SUMO substrates not isolable by classical chromatography techniques.


Sujet(s)
Protéines I-kappa B/isolement et purification , Sous-unité p50 de NF-kappa B/isolement et purification , Protéine SUMO-1/métabolisme , Facteur de transcription RelA/isolement et purification , Domaine catalytique , Chromatographie d'affinité , Électrophorèse sur gel de polyacrylamide , Escherichia coli/génétique , Escherichia coli/métabolisme , Vecteurs génétiques/génétique , Vecteurs génétiques/métabolisme , Humains , Protéines I-kappa B/génétique , Protéines I-kappa B/métabolisme , Inhibiteur alpha de NF-KappaB , Sous-unité p50 de NF-kappa B/génétique , Sous-unité p50 de NF-kappa B/métabolisme , Cartographie d'interactions entre protéines , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Protéine SUMO-1/génétique , Sumoylation , Facteur de transcription RelA/génétique , Facteur de transcription RelA/métabolisme , Ultrafiltration
20.
J Struct Biol ; 174(1): 245-51, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-20974256

RÉSUMÉ

MED25 (ARC92/ACID1) is a 747 residues subunit specific to higher eukaryote Mediator complex, an essential component of the RNA polymerase II general transcriptional machinery. MED25 is a target of the Herpes simplex virus transactivator protein VP16. MED25 interacts with VP16 through a central MED25 PTOV (Prostate tumour overexpressed)/ACID (Activator interacting domain) domain of unknown structure. As a first step towards understanding the mechanism of recruitment of transactivation domains by MED25, we report here the NMR structure of the MED25 ACID domain. The domain architecture consists of a closed ß-barrel with seven strands (Β1-Β7) and three α-helices (H1-H3), an architecture showing similarities to that of the SPOC (Spen paralog and ortholog C-terminal domain) domain-like superfamily. Preliminary NMR chemical shift mapping showed that VP16 H2 (VP16C) interacts with MED25 ACID through one face of the ß-barrel, defined by strands B4-B7-B6.


Sujet(s)
Complexe médiateur/composition chimique , Résonance magnétique nucléaire biomoléculaire/méthodes , Séquence d'acides aminés , Protéine Vmw65 de l'herpesvirus humain/métabolisme , Humains , Modèles moléculaires , Données de séquences moléculaires , Liaison aux protéines , Structure secondaire des protéines , Structure tertiaire des protéines , Similitude de séquences d'acides aminés , Similitude structurale de protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE