Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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.
Sci Rep ; 9(1): 13713, 2019 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-31548626

RESUMO

Use of microbes to produce liquid transportation fuels is not yet economically viable. A key point to reduce production costs is the design a cell factory that combines the continuous production of drop-in fuel molecules with the ability to recover products from the cell culture at low cost. Medium-chain hydrocarbons seem ideal targets because they can be produced from abundant fatty acids and, due to their volatility, can be easily collected in gas phase. However, pathways used to produce hydrocarbons from fatty acids require two steps, low efficient enzymes and/or complex electron donors. Recently, a new hydrocarbon-forming route involving a single enzyme called fatty acid photodecarboxylase (FAP) was discovered in microalgae. Here, we show that in illuminated E. coli cultures coexpression of FAP and a medium-chain fatty acid thioesterase results in continuous release of volatile hydrocarbons. Maximum hydrocarbon productivity was reached under low/medium light while higher irradiance resulted in decreased amounts of FAP. It was also found that the production rate of hydrocarbons was constant for at least 5 days and that 30% of total hydrocarbons could be collected in the gas phase of the culture. This work thus demonstrates that the photochemistry of the FAP can be harnessed to design a simple cell factory that continuously produces hydrocarbons easy to recover and in pure form.


Assuntos
Biocombustíveis , Ácidos Graxos/metabolismo , Hidrocarbonetos/metabolismo , Microalgas/metabolismo , Escherichia coli/metabolismo , Luz
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.
J Mol Biol ; 430(3): 297-309, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29223729

RESUMO

The type VI secretion system (T6SS) is a specialized macromolecular complex dedicated to the delivery of protein effectors into both eukaryotic and bacterial cells. The general mechanism of action of the T6SS is similar to the injection of DNA by contractile bacteriophages. The cytoplasmic portion of the T6SS is evolutionarily, structurally and functionally related to the phage tail complex. It is composed of an inner tube made of stacked Hcp hexameric rings, engulfed within a sheath and built on a baseplate. This sheath undergoes cycles of extension and contraction, and the current model proposes that the sheath contraction propels the inner tube toward the target cell for effector delivery. The sheath comprises two subunits: TssB and TssC that polymerize under an extended conformation. Here, we show that isolated TssB forms trimers, and we report the crystal structure of a C-terminal fragment of TssB. This fragment comprises a long helix followed by a helical hairpin that presents surface-exposed charged residues. Site-directed mutagenesis coupled to functional assay further showed that these charges are required for proper assembly of the sheath. Positioning of these residues in the extended T6SS sheath structure suggests that they may mediate contacts with the baseplate.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/química , Sistemas de Secreção Tipo VI/química , Cristalografia por Raios X , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Multimerização Proteica
5.
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
6.
Science ; 357(6354): 903-907, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860382

RESUMO

Although many organisms capture or respond to sunlight, few enzymes are known to be driven by light. Among these are DNA photolyases and the photosynthetic reaction centers. Here, we show that the microalga Chlorella variabilis NC64A harbors a photoenzyme that acts in lipid metabolism. This enzyme belongs to an algae-specific clade of the glucose-methanol-choline oxidoreductase family and catalyzes the decarboxylation of free fatty acids to n-alkanes or -alkenes in response to blue light. Crystal structure of the protein reveals a fatty acid-binding site in a hydrophobic tunnel leading to the light-capturing flavin adenine dinucleotide (FAD) cofactor. The decarboxylation is initiated through electron abstraction from the fatty acid by the photoexcited FAD with a quantum yield >80%. This photoenzyme, which we name fatty acid photodecarboxylase, may be useful in light-driven, bio-based production of hydrocarbons.


Assuntos
Alcanos/metabolismo , Alcenos/metabolismo , Biocatálise , Carboxiliases/metabolismo , Chlorella/enzimologia , Ácidos Graxos/metabolismo , Oxirredutases/metabolismo , Proteínas de Plantas/metabolismo , Carboxiliases/química , Carboxiliases/classificação , Carboxiliases/efeitos da radiação , Flavina-Adenina Dinucleotídeo/metabolismo , Luz , Metabolismo dos Lipídeos , Oxirredutases/química , Oxirredutases/classificação , Oxirredutases/efeitos da radiação , Processos Fotoquímicos , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/classificação , Proteínas de Plantas/efeitos da radiação
7.
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
8.
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
9.
Sci Rep ; 6: 34405, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27698444

RESUMO

The Type VI secretion system (T6SS) is a versatile machine that delivers toxins into either eukaryotic or bacterial cells. It thus represents a key player in bacterial pathogenesis and inter-bacterial competition. Schematically, the T6SS can be viewed as a contractile tail structure anchored to the cell envelope. The contraction of the tail sheath propels the inner tube loaded with effectors towards the target cell. The components of the contracted tail sheath are then recycled by the ClpV AAA+ ATPase for a new cycle of tail elongation. The T6SS is widespread in Gram-negative bacteria and most of their genomes carry several copies of T6SS gene clusters, which might be activated in different conditions. Here, we show that the ClpV ATPases encoded within the two T6SS gene clusters of enteroaggregative Escherichia coli are not interchangeable and specifically participate to the activity of their cognate T6SS. Here we show that this specificity is dictated by interaction between the ClpV N-terminal domains and the N-terminal helices of their cognate TssC1 proteins. We also present the crystal structure of the ClpV1 N-terminal domain, alone or in complex with the TssC1 N-terminal peptide, highlighting the commonalities and diversities in the recruitment of ClpV to contracted sheaths.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Família Multigênica , Sistemas de Secreção Tipo VI/química , Adenosina Trifosfatases/genética , Cristalografia por Raios X , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Domínios Proteicos , Sistemas de Secreção Tipo VI/genética
10.
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
11.
mBio ; 7(1): e01781-15, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26814179

RESUMO

UNLABELLED: The Gram-positive bacterium Lactococcus lactis, used for the production of cheeses and other fermented dairy products, falls victim frequently to fortuitous infection by tailed phages. The accompanying risk of dairy fermentation failures in industrial facilities has prompted in-depth investigations of these phages. Lactococcal phage Tuc2009 possesses extensive genomic homology to phage TP901-1. However, striking differences in the baseplate-encoding genes stimulated our interest in solving the structure of this host's adhesion device. We report here the X-ray structures of phage Tuc2009 receptor binding protein (RBP) and of a "tripod" assembly of three baseplate components, BppU, BppA, and BppL (the RBP). These structures made it possible to generate a realistic atomic model of the complete Tuc2009 baseplate that consists of an 84-protein complex: 18 BppU, 12 BppA, and 54 BppL proteins. The RBP head domain possesses a different fold than those of phages p2, TP901-1, and 1358, while the so-called "stem" and "neck" domains share structural features with their equivalents in phage TP901-1. The BppA module interacts strongly with the BppU N-terminal domain. Unlike other characterized lactococcal phages, Tuc2009 baseplate harbors two different carbohydrate recognition sites: one in the bona fide RBP head domain and the other in BppA. These findings represent a major step forward in deciphering the molecular mechanism by which Tuc2009 recognizes its saccharidic receptor(s) on its host. IMPORTANCE: Understanding how siphophages infect Lactococcus lactis is of commercial importance as they cause milk fermentation failures in the dairy industry. In addition, such knowledge is crucial in a general sense in order to understand how viruses recognize their host through protein-glycan interactions. We report here the lactococcal phage Tuc2009 receptor binding protein (RBP) structure as well as that of its baseplate. The RBP head domain has a different fold than those of phages p2, TP901-1, and 1358, while the so-called "stem" and "neck" share the fold characteristics also found in the equivalent baseplate proteins of phage TP901-1. The baseplate structure contains, in contrast to other characterized lactococcal phages, two different carbohydrate binding modules that may bind different motifs of the host's surface polysaccharide.


Assuntos
Bacteriófagos/química , Metabolismo dos Carboidratos , Lactococcus lactis/virologia , Proteínas da Cauda Viral/química , Proteínas da Cauda Viral/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Complexos Multiproteicos/química , Ligação Proteica , Conformação Proteica , Siphoviridae/química
12.
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
13.
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
14.
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
15.
J Mol Biol ; 426(22): 3757-3772, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25219509

RESUMO

Cutinases belong to the α/ß-hydrolase fold family of enzymes and degrade cutin and various esters, including triglycerides, phospholipids and galactolipids. Cutinases are able to degrade aggregated and soluble substrates because, in contrast with true lipases, they do not have a lid covering their catalytic machinery. We report here the structure of a cutinase from the fungus Trichoderma reesei (Tr) in native and inhibitor-bound conformations, along with its enzymatic characterization. A rare characteristic of Tr cutinase is its optimal activity at acidic pH. Furthermore, Tr cutinase, in contrast with classical cutinases, possesses a lid covering its active site and requires the presence of detergents for activity. In addition to the presence of the lid, the core of the Tr enzyme is very similar to other cutinase cores, with a central five-stranded ß-sheet covered by helices on either side. The catalytic residues form a catalytic triad involving Ser164, His229 and Asp216 that is covered by the two N-terminal helices, which form the lid. This lid opens in the presence of surfactants, such as ß-octylglucoside, and uncovers the catalytic crevice, allowing a C11Y4 phosphonate inhibitor to bind to the catalytic serine. Taken together, these results reveal Tr cutinase to be a member of a new group of lipolytic enzymes resembling cutinases but with kinetic and structural features of true lipases and a heightened specificity for long-chain triglycerides.


Assuntos
Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/metabolismo , Lipase/metabolismo , Trichoderma/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Domínio Catalítico , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Cinética , Lipólise , Modelos Moleculares , Dados de Sequência Molecular , Organofosfonatos/metabolismo , Conformação Proteica , Homologia de Sequência de Aminoácidos
16.
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
17.
PLoS One ; 9(2): e86918, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551044

RESUMO

The type VI secretion system (T6SS) is a widespread machine used by bacteria to control their environment and kill or disable bacterial species or eukaryotes through toxin injection. The T6SS comprises a central tube formed of stacked hexamers of hemolysin co-regulated proteins (Hcp) and terminated by a trimeric valine-glycine repeat protein G (VgrG) component, the cell puncturing device. A contractile tail sheath, formed by the TssB and TssC proteins, surrounds this tube. This syringe-like machine has been compared to an inverted phage, as both Hcp and VgrG share structural homology with tail components of Caudovirales. Here we solved the crystal structure of a tryptophan-substituted double mutant of Hcp1 from enteroaggregative Escherichia coli and compared it to the structures of other Hcps. Interestingly, we observed that the purified Hcp native protein is unable to form tubes in vitro. To better understand the rationale for observation, we measured the affinity of Hcp1 hexamers with themselves by surface plasmon resonance. The intra-hexamer interaction is weak, with a KD value of 7.2 µM. However, by engineering double cysteine mutants at defined positions, tubes of Hcp1 gathering up to 15 stacked hexamers formed in oxidative conditions. These results, together with those available in the literature regarding TssB and TssC, suggest that assembly of the T6SS tube differs significantly from that of Sipho- or Myoviridae.


Assuntos
Sistemas de Secreção Bacterianos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Cromatografia em Gel , Cristalografia por Raios X , Dissulfetos/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Luz , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ligação Proteica , Multimerização Proteica , Espalhamento de Radiação , Alinhamento de Sequência , Homologia Estrutural de Proteína , Ressonância de Plasmônio de Superfície
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...