Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Plant Physiol ; 186(3): 1455-1472, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-33856460

RESUMO

Fatty acid photodecarboxylase (FAP) is one of the few enzymes that require light for their catalytic cycle (photoenzymes). FAP was first identified in the microalga Chlorella variabilis NC64A, and belongs to an algae-specific subgroup of the glucose-methanol-choline oxidoreductase family. While the FAP from C. variabilis and its Chlamydomonas reinhardtii homolog CrFAP have demonstrated in vitro activities, their activities and physiological functions have not been studied in vivo. Furthermore, the conservation of FAP activity beyond green microalgae remains hypothetical. Here, using a C. reinhardtii FAP knockout line (fap), we showed that CrFAP is responsible for the formation of 7-heptadecene, the only hydrocarbon of this alga. We further showed that CrFAP was predominantly membrane-associated and that >90% of 7-heptadecene was recovered in the thylakoid fraction. In the fap mutant, photosynthetic activity was not affected under standard growth conditions, but was reduced after cold acclimation when light intensity varied. A phylogenetic analysis that included sequences from Tara Ocean identified almost 200 putative FAPs and indicated that FAP was acquired early after primary endosymbiosis. Within Bikonta, FAP was retained in secondary photosynthetic endosymbiosis lineages but absent from those that lost the plastid. Characterization of recombinant FAPs from various algal genera (Nannochloropsis, Ectocarpus, Galdieria, Chondrus) provided experimental evidence that FAP photochemical activity was present in red and brown algae, and was not limited to unicellular species. These results thus indicate that FAP was conserved during the evolution of most algal lineages where photosynthesis was retained, and suggest that its function is linked to photosynthetic membranes.


Assuntos
Carboxiliases/metabolismo , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Ácidos Graxos/metabolismo , Microalgas/metabolismo , Processos Fotoquímicos , Tilacoides/metabolismo , Ácidos Graxos/genética , Regulação da Expressão Gênica de Plantas , Genes de Plantas , Variação Genética , Genótipo , Luz , Microalgas/genética , Mutação , Tilacoides/genética
2.
Proc Natl Acad Sci U S A ; 114(45): 12063-12068, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29078388

RESUMO

The cytochrome (cyt) b6f complex and Stt7 kinase regulate the antenna sizes of photosystems I and II through state transitions, which are mediated by a reversible phosphorylation of light harvesting complexes II, depending on the redox state of the plastoquinone pool. When the pool is reduced, the cyt b6f activates the Stt7 kinase through a mechanism that is still poorly understood. After random mutagenesis of the chloroplast petD gene, coding for subunit IV of the cyt b6f complex, and complementation of a ΔpetD host strain by chloroplast transformation, we screened for impaired state transitions in vivo by chlorophyll fluorescence imaging. We show that residues Asn122, Tyr124, and Arg125 in the stromal loop linking helices F and G of cyt b6f subunit IV are crucial for state transitions. In vitro reconstitution experiments with purified cyt b6f and recombinant Stt7 kinase domain show that cyt b6f enhances Stt7 autophosphorylation and that the Arg125 residue is directly involved in this process. The peripheral stromal structure of the cyt b6f complex had, until now, no reported function. Evidence is now provided of a direct interaction with Stt7 on the stromal side of the membrane.


Assuntos
Chlamydomonas/metabolismo , Complexo Citocromos b6f/metabolismo , Proteínas Quinases/metabolismo , Clorofila/metabolismo , Cloroplastos/metabolismo , Complexos de Proteínas Captadores de Luz/metabolismo , Oxirredução , Fosforilação/fisiologia , Complexo de Proteína do Fotossistema I/metabolismo , Complexo de Proteína do Fotossistema II/metabolismo , Plastoquinona/metabolismo
3.
Plant Physiol ; 177(4): 1639-1649, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29976836

RESUMO

Some microalgae, such as Chlamydomonas reinhardtii, harbor a highly flexible photosynthetic apparatus capable of using different electron acceptors, including carbon dioxide (CO2), protons, or oxygen (O2), allowing survival in diverse habitats. During anaerobic induction of photosynthesis, molecular O2 is produced at photosystem II, while at the photosystem I acceptor side, the reduction of protons into hydrogen (H2) by the plastidial [FeFe]-hydrogenases primes CO2 fixation. Although the interaction between H2 production and CO2 fixation has been studied extensively, their interplay with O2 produced by photosynthesis has not been considered. By simultaneously measuring gas exchange and chlorophyll fluorescence, we identified an O2 photoreduction mechanism that functions during anaerobic dark-to-light transitions and demonstrate that flavodiiron proteins (Flvs) are the major players involved in light-dependent O2 uptake. We further show that Flv-mediated O2 uptake is critical for the rapid induction of CO2 fixation but is not involved in the creation of the micro-oxic niches proposed previously to protect the [FeFe]-hydrogenase from O2 By studying a mutant lacking both hydrogenases (HYDA1 and HYDA2) and both Flvs (FLVA and FLVB), we show that the induction of photosynthesis is strongly delayed in the absence of both sets of proteins. Based on these data, we propose that Flvs are involved in an important intracellular O2 recycling process, which acts as a relay between H2 production and CO2 fixation.


Assuntos
Dióxido de Carbono/metabolismo , Chlamydomonas reinhardtii/fisiologia , Hidrogênio/metabolismo , Oxigênio/metabolismo , Proteínas de Plantas/metabolismo , Anaerobiose , Clorofila/metabolismo , Flavoproteínas/genética , Flavoproteínas/metabolismo , Fluorescência , Hidrogenase/metabolismo , Mutação , Processos Fotoquímicos , Fotossíntese/fisiologia
4.
Plant J ; 90(2): 358-371, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28142200

RESUMO

Peroxisomes are thought to have played a key role in the evolution of metabolic networks of photosynthetic organisms by connecting oxidative and biosynthetic routes operating in different compartments. While the various oxidative pathways operating in the peroxisomes of higher plants are fairly well characterized, the reactions present in the primitive peroxisomes (microbodies) of algae are poorly understood. Screening of a Chlamydomonas insertional mutant library identified a strain strongly impaired in oil remobilization and defective in Cre05.g232002 (CrACX2), a gene encoding a member of the acyl-CoA oxidase/dehydrogenase superfamily. The purified recombinant CrACX2 expressed in Escherichia coli catalyzed the oxidation of fatty acyl-CoAs into trans-2-enoyl-CoA and produced H2 O2 . This result demonstrated that CrACX2 is a genuine acyl-CoA oxidase, which is responsible for the first step of the peroxisomal fatty acid (FA) ß-oxidation spiral. A fluorescent protein-tagging study pointed to a peroxisomal location of CrACX2. The importance of peroxisomal FA ß-oxidation in algal physiology was shown by the impact of the mutation on FA turnover during day/night cycles. Moreover, under nitrogen depletion the mutant accumulated 20% more oil than the wild type, illustrating the potential of ß-oxidation mutants for algal biotechnology. This study provides experimental evidence that a plant-type FA ß-oxidation involving H2 O2 -producing acyl-CoA oxidation activity has already evolved in the microbodies of the unicellular green alga Chlamydomonas reinhardtii.


Assuntos
Acil-CoA Oxidase/metabolismo , Chlamydomonas/enzimologia , Chlamydomonas/metabolismo , Peroxissomos/metabolismo , Chlamydomonas/genética , Peróxido de Hidrogênio/metabolismo , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Nitrogênio/metabolismo , Oxirredução
5.
Plant Physiol ; 174(3): 1825-1836, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28487478

RESUMO

During oxygenic photosynthesis, the reducing power generated by light energy conversion is mainly used to reduce carbon dioxide. In bacteria and archae, flavodiiron (Flv) proteins catalyze O2 or NO reduction, thus protecting cells against oxidative or nitrosative stress. These proteins are found in cyanobacteria, mosses, and microalgae, but have been lost in angiosperms. Here, we used chlorophyll fluorescence and oxygen exchange measurement using [18O]-labeled O2 and a membrane inlet mass spectrometer to characterize Chlamydomonas reinhardtii flvB insertion mutants devoid of both FlvB and FlvA proteins. We show that Flv proteins are involved in a photo-dependent electron flow to oxygen, which drives most of the photosynthetic electron flow during the induction of photosynthesis. As a consequence, the chlorophyll fluorescence patterns are strongly affected in flvB mutants during a light transient, showing a lower PSII operating yield and a slower nonphotochemical quenching induction. Photoautotrophic growth of flvB mutants was indistinguishable from the wild type under constant light, but severely impaired under fluctuating light due to PSI photo damage. Remarkably, net photosynthesis of flv mutants was higher than in the wild type during the initial hour of a fluctuating light regime, but this advantage vanished under long-term exposure, and turned into PSI photo damage, thus explaining the marked growth retardation observed in these conditions. We conclude that the C. reinhardtii Flv participates in a Mehler-like reduction of O2, which drives a large part of the photosynthetic electron flow during a light transient and is thus critical for growth under fluctuating light regimes.


Assuntos
Chlamydomonas/metabolismo , Chlamydomonas/efeitos da radiação , Flavoproteínas/metabolismo , Luz , Oxigênio/metabolismo , Chlamydomonas/genética , Chlamydomonas/crescimento & desenvolvimento , Clorofila/metabolismo , Transporte de Elétrons , Fluorescência , Espectrometria de Massas , Mutação/genética , Oxirredução , Paraquat/farmacologia , Fotossíntese/efeitos da radiação , Complexo de Proteína do Fotossistema I/metabolismo , Complexo de Proteína do Fotossistema II/metabolismo
6.
J Biol Chem ; 291(32): 16816-27, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27268053

RESUMO

Glycerophosphodiester phosphodiesterases (GDPDs; EC 3.1.4.46) typically hydrolyze glycerophosphodiesters to sn-glycerol 3-phosphate (Gro3P) and their corresponding alcohol during patho/physiological processes in bacteria and eukaryotes. GDPD(-like) domains were identified in the structural particle of bacterial viruses (bacteriophages) specifically infecting Gram-positive bacteria. The GDPD of phage 17 (Ld17; GDPDLd17), representative of the group b Lactobacillus delbrueckii subsp. bulgaricus (Ldb)-infecting bacteriophages, was shown to hydrolyze, besides the simple glycerophosphodiester, two complex surface-associated carbohydrates of the Ldb17 cell envelope: the Gro3P decoration of the major surface polysaccharide d-galactan and the oligo(glycerol phosphate) backbone of the partially glycosylated cell wall teichoic acid, a minor Ldb17 cell envelope component. Degradation of cell wall teichoic acid occurs according to an exolytic mechanism, and Gro3P substitution is presumed to be inhibitory for GDPDLd17 activity. The presence of the GDPDLd17 homotrimer in the viral baseplate structure involved in phage-host interaction together with the dependence of native GDPD activity, adsorption, and efficiency of plating of Ca(2+) ions supports a role for GDPDLd17 activity during phage adsorption and/or phage genome injection. In contrast to GDPDLd17, we could not identify any enzymatic activity for the GDPD-like domain in the neck passage structure of phage 340, a 936-type Lactococcus lactis subsp. lactis bacteriophage.


Assuntos
Bacteriófagos/enzimologia , Lactobacillus delbrueckii/virologia , Diester Fosfórico Hidrolases/metabolismo , Proteínas Virais/metabolismo , Bacteriófagos/genética , Lactobacillus delbrueckii/metabolismo , Diester Fosfórico Hidrolases/genética , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo , Proteínas Virais/genética
7.
Mol Microbiol ; 99(6): 1099-118, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26714038

RESUMO

The Type VI secretion system (T6SS) is a multiprotein machine that delivers protein effectors in both prokaryotic and eukaryotic cells, allowing interbacterial competition and virulence. The mechanism of action of the T6SS requires the contraction of a sheath-like structure that propels a needle towards target cells, allowing the delivery of protein effectors. Here, we provide evidence that the entero-aggregative Escherichia coli Sci-1 T6SS is required to eliminate competitor bacteria. We further identify Tle1, a toxin effector encoded by this cluster and showed that Tle1 possesses phospholipase A1 and A2 activities required for the interbacterial competition. Self-protection of the attacker cell is secured by an outer membrane lipoprotein, Tli1, which binds Tle1 in a 1:1 stoichiometric ratio with nanomolar affinity, and inhibits its phospholipase activity. Tle1 is delivered into the periplasm of the prey cells using the VgrG1 needle spike protein as carrier. Further analyses demonstrate that the C-terminal extension domain of VgrG1, including a transthyretin-like domain, is responsible for the interaction with Tle1 and its subsequent delivery into target cells. Based on these results, we propose an additional mechanism of transport of T6SS effectors in which cognate effectors are selected by specific motifs located at the C-terminus of VgrG proteins.


Assuntos
Escherichia coli/metabolismo , Fosfolipases A1/metabolismo , Sistemas de Secreção Tipo VI/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Caenorhabditis elegans , Escherichia coli/patogenicidade , Modelos Moleculares , Família Multigênica , Fosfolipases A1/química , Fosfolipases A1/genética , Domínios Proteicos , Sistemas de Secreção Tipo VI/genética , Virulência
8.
Mol Microbiol ; 96(4): 875-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25708888

RESUMO

Each phage infects a limited number of bacterial strains through highly specific interactions of the receptor-binding protein (RBP) at the tip of phage tail and the receptor at the bacterial surface. Lactococcus lactis is covered with a thin polysaccharide pellicle (hexasaccharide repeating units), which is used by a subgroup of phages as a receptor. Using L. lactis and phage 1358 as a model, we investigated the interaction between the phage RBP and the pellicle hexasaccharide of the host strain. A core trisaccharide (TriS), derived from the pellicle hexasaccharide repeating unit, was chemically synthesised, and the crystal structure of the RBP/TriS complex was determined. This provided unprecedented structural details of RBP/receptor site-specific binding. The complete hexasaccharide repeating unit was modelled and found to aptly fit the extended binding site. The specificity observed in in vivo phage adhesion assays could be interpreted in view of the reported structure. Therefore, by combining synthetic carbohydrate chemistry, X-ray crystallography and phage plaquing assays, we suggest that phage adsorption results from distinct recognition of the RBP towards the core TriS or the remaining residues of the hexasacchride receptor. This study provides a novel insight into the adsorption process of phages targeting saccharides as their receptors.


Assuntos
Bacteriófagos/metabolismo , Polissacarídeos Bacterianos/metabolismo , Receptores Virais/metabolismo , Sítios de Ligação , Configuração de Carboidratos , Cristalografia por Raios X , Lactococcus lactis/metabolismo , Modelos Moleculares , Polissacarídeos Bacterianos/química , Ligação Proteica , Conformação Proteica , Receptores Virais/química , Trissacarídeos/síntese química
9.
Proc Natl Acad Sci U S A ; 110(15): E1371-9, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23530214

RESUMO

Lactococcal phages belong to a large family of Siphoviridae and infect Lactococcus lactis, a gram-positive bacterium used in commercial dairy fermentations. These phages are believed to recognize and bind specifically to pellicle polysaccharides covering the entire bacterium. The phage TP901-1 baseplate, located at the tip of the tail, harbors 18 trimeric receptor binding proteins (RBPs) promoting adhesion to a specific lactococcal strain. Phage TP901-1 adhesion does not require major conformational changes or Ca(2+), which contrasts other lactococcal phages. Here, we produced and characterized llama nanobodies raised against the purified baseplate and the Tal protein of phage TP901-1 as tools to dissect the molecular determinants of phage TP901-1 infection. Using a set of complementary techniques, surface plasmon resonance, EM, and X-ray crystallography in a hybrid approach, we identified binders to the three components of the baseplate, analyzed their affinity for their targets, and determined their epitopes as well as their functional impact on TP901-1 phage infectivity. We determined the X-ray structures of three nanobodies in complex with the RBP. Two of them bind to the saccharide binding site of the RBP and are able to fully neutralize TP901-1 phage infectivity, even after 15 passages. These results provide clear evidence for a practical use of nanobodies in circumventing lactococcal phages viral infection in dairy fermentation.


Assuntos
Lactococcus lactis/virologia , Anticorpos de Domínio Único/química , Siphoviridae/fisiologia , Proteínas da Cauda Viral/química , Animais , Especificidade de Anticorpos , Sítios de Ligação , Camelídeos Americanos , Cristalografia por Raios X , Epitopos/química , Fermentação , Microscopia Eletrônica , Modelos Moleculares , Conformação Molecular , Nanotecnologia , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície
10.
J Virol ; 88(12): 7005-15, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24719416

RESUMO

UNLABELLED: The Gram-positive bacterium Lactococcus lactis is used for the production of cheeses and other fermented dairy products. Accidental infection of L. lactis cells by virulent lactococcal tailed phages is one of the major risks of fermentation failures in industrial dairy factories. Lactococcal phage 1358 possesses a host range limited to a few L. lactis strains and strong genomic similarities to Listeria phages. We report here the X-ray structures of phage 1358 receptor binding protein (RBP) in complex with monosaccharides. Each monomer of its trimeric RBP is formed of two domains: a "shoulder" domain linking the RBP to the rest of the phage and a jelly roll fold "head/host recognition" domain. This domain harbors a saccharide binding crevice located in the middle of a monomer. Crystal structures identified two sites at the RBP surface, ∼8 Šfrom each other, one accommodating a GlcNAc monosaccharide and the other accommodating a GlcNAc or a glucose 1-phosphate (Glc1P) monosaccharide. GlcNAc and GlcNAc1P are components of the polysaccharide pellicle that we identified at the cell surface of L. lactis SMQ-388, the host of phage 1358. We therefore modeled a galactofuranose (Galf) sugar bridging the two GlcNAc saccharides, suggesting that the trisaccharidic motif GlcNAc-Galf-GlcNAc (or Glc1P) might be common to receptors of genetically distinct lactococcal phages p2, TP091-1, and 1358. Strain specificity might therefore be elicited by steric clashes induced by the remaining components of the pellicle hexasaccharide. Taken together, these results provide a first insight into the molecular mechanism of host receptor recognition by lactococcal phages. IMPORTANCE: Siphophages infecting the Gram-positive bacterium Lactococcus lactis are sources of milk fermentation failures in the dairy industry. We report here the structure of the pellicle polysaccharide from L. lactis SMQ-388, the specific host strain of phage 1358. We determined the X-ray structures of the lytic lactococcal phage 1358 receptor binding protein (RBP) in complex with monosaccharides. The positions and nature of monosaccharides bound to the RBP are in agreement with the pellicle structure and suggest a general binding mode of lactococcal phages to their pellicle saccharidic receptor.


Assuntos
Proteínas de Bactérias/química , Bacteriófagos/metabolismo , Parede Celular/virologia , Lactococcus lactis/virologia , Receptores Virais/química , Proteínas Virais/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriófagos/química , Bacteriófagos/genética , Parede Celular/química , Parede Celular/genética , Parede Celular/metabolismo , Cristalografia por Raios X , Lactococcus lactis/química , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Modelos Moleculares , Ligação Proteica , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
J Biol Inorg Chem ; 20(5): 905-19, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26070361

RESUMO

Calmodulin (CaM) is an essential Ca(II)-dependent regulator of cell physiology. To understand its interaction with Ca(II) at a molecular level, it is essential to examine Ca(II) binding at each site of the protein, even if it is challenging to estimate the site-specific binding properties of the interdependent CaM-binding sites. In this study, we evaluated the site-specific Ca(II)-binding affinity of sites I and II of the N-terminal domain by combining site-directed mutagenesis and spectrofluorimetry. The mutations had very low impact on the protein structure and stability. We used these binding constants to evaluate the inter-site cooperativity energy and compared it with its lower limit value usually reported in the literature. We found that site I affinity for Ca(II) was 1.5 times that of site II and that cooperativity induced an approximately tenfold higher affinity for the second Ca(II)-binding event, as compared to the first one. We further showed that insertion of a tryptophan at position 7 of site II binding loop significantly increased site II affinity for Ca(II) and the intra-domain cooperativity. ΔH and ΔS parameters were studied by isothermal titration calorimetry for Ca(II) binding to site I, site II and to the entire N-terminal domain. They showed that calcium binding is mainly entropy driven for the first and second binding events. These findings provide molecular information on the structure-affinity relationship of the individual sites of the CaM N-terminal domain and new perspectives for the optimization of metal ion binding by mutating the EF-hand loops sequences.


Assuntos
Cálcio/química , Calmodulina/química , Termodinâmica , Sequência de Aminoácidos , Sítios de Ligação , Calmodulina/genética , Calmodulina/isolamento & purificação , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Engenharia de Proteínas , Estrutura Terciária de Proteína
12.
Proc Natl Acad Sci U S A ; 109(23): 8954-8, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22611190

RESUMO

Phages of the Caudovirales order possess a tail that recognizes the host and ensures genome delivery upon infection. The X-ray structure of the approximately 1.8 MDa host adsorption device (baseplate) from the lactococcal phage TP901-1 shows that the receptor-binding proteins are pointing in the direction of the host, suggesting that this organelle is in a conformation ready for host adhesion. This result is in marked contrast with the lactococcal phage p2 situation, whose baseplate is known to undergo huge conformational changes in the presence of Ca(2+) to reach its active state. In vivo infection experiments confirmed these structural observations by demonstrating that Ca(2+) ions are required for host adhesion among p2-like phages (936-species) but have no influence on TP901-1-like phages (P335-species). These data suggest that these two families rely on diverse adhesion strategies which may lead to different signaling for genome release.


Assuntos
Caudovirales/genética , Modelos Moleculares , Proteínas da Cauda Viral/genética , Ligação Viral , Bacteriófago P2/genética , Cálcio/metabolismo , Cristalografia , Lactococcus lactis/virologia , Proteínas da Cauda Viral/química , Proteínas da Cauda Viral/metabolismo
13.
Mol Microbiol ; 89(1): 152-65, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23692331

RESUMO

Lipoproteins of temperate phage are a broad family of membrane proteins encoded in the lysogeny module of temperate phages. Expression of the ltp(TP-J34) gene of temperate Streptococcus thermophilus phage TP-J34 interferes with phage infection at the stage of triggering DNA release and injection into the cell. Here, we report the first structure of a superinfection exclusion protein. We have expressed and determined the X-ray structure of Ltp(TP-J34). The soluble domain of Ltp(TP-J34) is composed of a tandem of three-helix helix-turn-helix (HTH) domains exhibiting a highly negatively charged surface. By isolating mutants of lactococcal phage P008wt with reduced sensitivities to Ltp(TP-J34) and by genome sequencing of such mutants we obtained evidence supporting the notion that Ltp(TP-J34) targets the phage's tape measure protein (TMP) and blocks its insertion into the cytoplasmic membrane.


Assuntos
Lipoproteínas/química , Lipoproteínas/metabolismo , Fagos de Streptococcus/química , Proteínas Virais/química , Proteínas Virais/metabolismo , Cristalografia por Raios X , Lisogenia , Prófagos/química , Conformação Proteica , Streptococcus thermophilus/virologia
14.
J Virol ; 87(15): 8429-40, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23698314

RESUMO

Many phages employ a large heteropolymeric organelle located at the tip of the tail, termed the baseplate, for host recognition. Contrast electron microscopy (EM) of the lactococcal phage Tuc2009 baseplate and its host-binding subunits, the so-called tripods, allowed us to obtain a low-resolution structural image of this organelle. Structural comparisons between the baseplate of the related phage TP901-1 and that of Tuc2009 demonstrated that they are highly similar, except for the presence of an additional protein in the Tuc2009 baseplate (BppATuc2009), which is attached to the top of the Tuc2009 tripod structure. Recombinantly produced Tuc2009 or TP901-1 tripods were shown to bind specifically to their particular host cell surfaces and are capable of almost fully and specifically eliminating Tuc2009 or TP901-1 phage adsorption, respectively. In the case of Tuc2009, such adsorption-blocking ability was reduced in tripods that lacked BppATuc2009, indicating that this protein increases the binding specificity and/or affinity of the Tuc2009 tripod to its host receptor.


Assuntos
Bacteriófagos/fisiologia , Bacteriófagos/ultraestrutura , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Ligação Viral , Sequência de Aminoácidos , Imageamento Tridimensional , Lactococcus/virologia , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Dados de Sequência Molecular , Alinhamento de Sequência , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura
15.
PLoS Pathog ; 7(11): e1002386, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22102820

RESUMO

Type VI secretion systems (T6SS) are trans-envelope machines dedicated to the secretion of virulence factors into eukaryotic or prokaryotic cells, therefore required for pathogenesis and/or for competition towards neighboring bacteria. The T6SS apparatus resembles the injection device of bacteriophage T4, and is anchored to the cell envelope through a membrane complex. This membrane complex is composed of the TssL, TssM and TagL inner membrane anchored proteins and of the TssJ outer membrane lipoprotein. Here, we report the crystal structure of the enteroaggregative Escherichia coli Sci1 TssJ lipoprotein, a two four-stranded ß-sheets protein that exhibits a transthyretin fold with an additional α-helical domain and a protruding loop. We showed that TssJ contacts TssM through this loop since a loop depleted mutant failed to interact with TssM in vitro or in vivo. Biophysical analysis of TssM and TssJ-TssM interaction suggest a structural model of the membrane-anchored outer shell of T6SS. Collectively, our results provide an improved understanding of T6SS assembly and encourage structure-aided drug design of novel antimicrobials targeting T6SS.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Sistemas de Secreção Bacterianos , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Lipopeptídeos/química , Proteínas de Membrana/química , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/metabolismo , Lipopeptídeos/metabolismo , Proteínas de Membrana/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Virulência/química , Fatores de Virulência/metabolismo
16.
J Biol Chem ; 286(15): 13583-602, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21317293

RESUMO

The Henipavirus genome is encapsidated by the nucleoprotein (N) within a helical nucleocapsid that recruits the polymerase complex via the phosphoprotein (P). In a previous study, we reported that in henipaviruses, the N-terminal domain of the phosphoprotein and the C-terminal domain of the nucleoprotein (N(TAIL)) are both intrinsically disordered. Here we show that Henipavirus N(TAIL) domains are also disordered in the context of full-length nucleoproteins. We also report the cloning, purification, and characterization of the C-terminal X domains (P(XD)) of Henipavirus phosphoproteins. Using isothermal titration calorimetry, we show that N(TAIL) and P(XD) form a 1:1 stoichiometric complex that is stable under NaCl concentrations as high as 1 M and has a K(D) in the µM range. Using far-UV circular dichroism and nuclear magnetic resonance, we show that P(XD) triggers an increase in the α-helical content of N(TAIL). Using fluorescence spectroscopy, we show that P(XD) has no impact on the chemical environment of a Trp residue introduced at position 527 of the Henipavirus N(TAIL) domain, thus arguing for the lack of stable contacts between the C termini of N(TAIL) and P(XD). Finally, we present a tentative structural model of the N(TAIL)-P(XD) interaction in which a short, order-prone region of N(TAIL) (α-MoRE; amino acids 473-493) adopts an α-helical conformation and is embedded between helices α2 and α3 of P(XD), leading to a relatively small interface dominated by hydrophobic contacts. The present results provide the first detailed experimental characterization of the N-P interaction in henipaviruses and designate the N(TAIL)-P(XD) interaction as a valuable target for rational antiviral approaches.


Assuntos
Henipavirus/química , Modelos Moleculares , Nucleoproteínas/química , Fosfoproteínas/química , Proteínas Virais/química , Henipavirus/genética , Nucleoproteínas/genética , Fosfoproteínas/genética , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Virais/genética
17.
Proc Natl Acad Sci U S A ; 106(50): 21155-60, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19934032

RESUMO

Acidianus filamentous virus 1 (AFV1), a member of the Lipothrixviridae family, infects the hyperthermophilic, acidophilic crenarchaeaon Acidianus hospitalis. The virion, covered with a lipidic outer shell, is 9,100-A long and contains a 20.8-kb linear dsDNA genome. We have identified the two major coat proteins of the virion (MCPs; 132 and 140 amino acids). They bind DNA and form filaments when incubated with linear dsDNA. A C-terminal domain is identified in their crystal structure with a four-helix-bundle fold. In the topological model of the virion filament core, the genomic dsDNA superhelix wraps around the AFV1-132 basic protein, and the AFV1-140 basic N terminus binds genomic DNA, while its lipophilic C-terminal domain is imbedded in the lipidic outer shell. The four-helix bundle fold of the MCPs from AFV1 is identical to that of the coat protein (CP) of Sulfolobus islandicus rod-shaped virus (SIRV), a member of the Rudiviridae family. Despite low sequence identity between these proteins, their high degree of structural similarity suggests that they could have derived from a common ancestor and could thus define an yet undescribed viral lineage.


Assuntos
Proteínas do Capsídeo/química , Lipothrixviridae/química , Dobramento de Proteína , Acidianus/virologia , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Genoma Viral , Lipothrixviridae/genética , Dados de Sequência Molecular , Filogenia , Estrutura Secundária de Proteína , Homologia Estrutural de Proteína , Sulfolobus/química
18.
J Struct Biol ; 172(1): 75-84, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20153432

RESUMO

We report here the characterization of several large structural protein complexes forming the baseplates (or part of them) of Siphoviridae phages infecting Lactococcus lactis: TP901-1, Tuc2009 and p2. We revisited a "block cloning" expression strategy and extended this approach to genomic fragments encoding proteins whose interacting partners have not yet been clearly identified. Biophysical characterization of some of these complexes using circular dichroism and size exclusion chromatography, coupled with on-line light scattering and refractometry, demonstrated that the over-produced recombinant proteins interact with each other to form large (up to 1.9MDa) and stable baseplate assemblies. Some of these complexes were characterized by electron microscopy confirming their structural homogeneity as well as providing a picture of their overall molecular shapes and symmetry. Finally, using these results, we were able to highlight similarities and differences with the well characterized much larger baseplate of the myophage T4.


Assuntos
Lactococcus lactis/virologia , Proteínas Recombinantes/metabolismo , Siphoviridae/metabolismo , Proteínas Virais/metabolismo , Cromatografia em Gel , Dicroísmo Circular , Escherichia coli/genética , Microscopia Eletrônica , Peso Molecular , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/ultraestrutura , Siphoviridae/genética , Siphoviridae/ultraestrutura , Soluções , Proteínas Virais/química , Proteínas Virais/genética
19.
J Biol Chem ; 284(44): 30718-26, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19740746

RESUMO

Combinatorial libraries of designed ankyrin repeat proteins (DARPins) have been proven to be a valuable source of specific binding proteins, as they can be expressed at very high levels and are very stable. We report here the selection of DARPins directed against a macromolecular multiprotein complex, the baseplate BppUxBppL complex of the lactococcal phage TP901-1. Using ribosome display, we selected several DARPins that bound specifically to the tip of the receptor-binding protein (RBP, the BppL trimer). The three selected DARPins display high specificity and affinity in the low nanomolar range and bind with a stoichiometry of one DARPin per BppL trimer. The crystal structure of a DARPin complexed with the RBP was solved at 2.1 A resolution. The DARPinxRBP interface is of the concave (DARPin)-convex (RBP) type, typical of other DARPin protein complexes and different from what is observed with a camelid VHH domain, which penetrates the phage p2 RBP inter-monomer interface. Finally, phage infection assays demonstrated that TP901-1 infection of Lactococcus lactis cells was inhibited by each of the three selected DARPins. This study provides proof of concept for the possible use of DARPins to circumvent viral infection. It also provides support for the use of DARPins in co-crystallization, due to their rigidity and their ability to provide multiple crystal contacts.


Assuntos
Repetição de Anquirina/genética , Antivirais/química , Bacteriófagos/química , Lactococcus lactis/virologia , Proteínas Virais/química , Viroses/prevenção & controle , Sítios de Ligação , Cristalografia por Raios X , Complexos Multiproteicos/química , Biblioteca de Peptídeos , Conformação Proteica , Engenharia de Proteínas , Receptores Virais/metabolismo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/metabolismo
20.
J Bacteriol ; 191(10): 3220-5, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19286807

RESUMO

Lactococcus lactis, a gram-positive bacterium widely used by the dairy industry to manufacture cheeses, is subject to infection by a diverse population of virulent phages. We have previously determined the structures of three receptor binding proteins (RBPs) from lactococcal phages TP901-1, p2, and bIL170, each of them having a distinct host range. Virulent phages p2 and bIL170 are classified within the 936 group, while the temperate phage TP901-1 is a member of the genetically distinct P335 polythetic group. These RBPs comprise three domains: the N-terminal domain, binding to the virion particle; a beta-helical linker domain; and the C-terminal domain, bearing the receptor binding site used for host recognition. Here, we have designed, expressed, and determined the structure of an RBP chimera in which the N-terminal and linker RBP domains of phage TP901-1 (P335) are fused to the C-terminal RBP domain of phage p2 (936). This chimera exhibits a stable structure that closely resembles the parental structures, while a slight displacement of the linker made RBP domain adaptation efficient. The receptor binding site is structurally indistinguishable from that of native p2 RBP and binds glycerol with excellent affinity.


Assuntos
Bacteriófagos/metabolismo , Cristalografia por Raios X/métodos , Lactococcus lactis/virologia , Proteínas Recombinantes de Fusão/química , Proteínas Virais/química , Sítios de Ligação , Peptídeo Hidrolases/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA