Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Acta Crystallogr F Struct Biol Commun ; 80(Pt 5): 98-106, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38699971

ABSTRACT

Molybdenum- or tungsten-dependent formate dehydrogenases have emerged as significant catalysts for the chemical reduction of CO2 to formate, with biotechnological applications envisaged in climate-change mitigation. The role of Met405 in the active site of Desulfovibrio vulgaris formate dehydrogenase AB (DvFdhAB) has remained elusive. However, its proximity to the metal site and the conformational change that it undergoes between the resting and active forms suggests a functional role. In this work, the M405S variant was engineered, which allowed the active-site geometry in the absence of methionine Sδ interactions with the metal site to be revealed and the role of Met405 in catalysis to be probed. This variant displayed reduced activity in both formate oxidation and CO2 reduction, together with an increased sensitivity to oxygen inactivation.


Subject(s)
Desulfovibrio vulgaris , Formate Dehydrogenases , Desulfovibrio vulgaris/enzymology , Desulfovibrio vulgaris/genetics , Formate Dehydrogenases/chemistry , Formate Dehydrogenases/genetics , Formate Dehydrogenases/metabolism , Catalytic Domain , Crystallography, X-Ray , Oxidation-Reduction , Models, Molecular , Formates/metabolism , Formates/chemistry , Carbon Dioxide/metabolism , Carbon Dioxide/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
Nat Chem Biol ; 20(1): 111-119, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37985883

ABSTRACT

Metal-dependent formate dehydrogenases reduce CO2 with high efficiency and selectivity, but are usually very oxygen sensitive. An exception is Desulfovibrio vulgaris W/Sec-FdhAB, which can be handled aerobically, but the basis for this oxygen tolerance was unknown. Here we show that FdhAB activity is controlled by a redox switch based on an allosteric disulfide bond. When this bond is closed, the enzyme is in an oxygen-tolerant resting state presenting almost no catalytic activity and very low formate affinity. Opening this bond triggers large conformational changes that propagate to the active site, resulting in high activity and high formate affinity, but also higher oxygen sensitivity. We present the structure of activated FdhAB and show that activity loss is associated with partial loss of the metal sulfido ligand. The redox switch mechanism is reversible in vivo and prevents enzyme reduction by physiological formate levels, conferring a fitness advantage during O2 exposure.


Subject(s)
Carbon Dioxide , Oxidoreductases , Carbon Dioxide/chemistry , Oxygen , Oxidation-Reduction , Catalytic Domain , Formates
3.
J Biol Chem ; 299(8): 105036, 2023 08.
Article in English | MEDLINE | ID: mdl-37442232

ABSTRACT

Arsenic contamination of groundwater is among one of the biggest health threats affecting millions of people in the world. There is an urgent need for efficient arsenic biosensors where the use of arsenic metabolizing enzymes can be explored. In this work, we have solved four crystal structures of arsenite oxidase (Aio) in complex with arsenic and antimony oxyanions and the structures determined correspond to intermediate states of the enzymatic mechanism. These structural data were complemented with density-functional theory calculations providing a unique view of the molybdenum active site at different time points that, together with mutagenesis data, enabled to clarify the enzymatic mechanism and the molecular determinants for the oxidation of As(III) to the less toxic As(V) species.


Subject(s)
Arsenic , Arsenites , Humans , Antimony , Oxidation-Reduction
4.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047219

ABSTRACT

Neisseria gonorrhoeae is an obligate human pathogenic bacterium responsible for gonorrhea, a sexually transmitted disease. The bacterial peroxidase, an enzyme present in the periplasm of this bacterium, detoxifies the cells against hydrogen peroxide and constitutes one of the primary defenses against exogenous and endogenous oxidative stress in this organism. The 38 kDa heterologously produced bacterial peroxidase was crystallized in the mixed-valence state, the active state, at pH 6.0, and the crystals were soaked with azide, producing the first azide-inhibited structure of this family of enzymes. The enzyme binds exogenous ligands such as cyanide and azide, which also inhibit the catalytic activity by coordinating the P heme iron, the active site, and competing with its substrate, hydrogen peroxide. The inhibition constants were estimated to be 0.4 ± 0.1 µM and 41 ± 5 mM for cyanide and azide, respectively. Imidazole also binds and inhibits the enzyme in a more complex mechanism by binding to P and E hemes, which changes the reduction potential of the latest heme. Based on the structures now reported, the catalytic cycle of bacterial peroxidases is revisited. The inhibition studies and the crystal structure of the inhibited enzyme comprise the first platform to search and develop inhibitors that target this enzyme as a possible new strategy against N. gonorrhoeae.


Subject(s)
Peroxidase , Peroxidases , Humans , Peroxidases/metabolism , Neisseria gonorrhoeae , Hydrogen Peroxide/metabolism , Azides/chemistry , Heme/metabolism
5.
Infect Immun ; 91(4): e0040522, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36877064

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that causes ocular and urogenital infections in humans. The ability of C. trachomatis to grow intracellularly in a pathogen-containing vacuole (known as an inclusion) depends on chlamydial effector proteins transported into the host cell by a type III secretion system. Among these effectors, several inclusion membrane proteins (Incs) insert in the vacuolar membrane. Here, we show that human cell lines infected by a C. trachomatis strain deficient for Inc CT288/CTL0540 (renamed IncM) displayed less multinucleation than when infected by IncM-producing strains (wild type or complemented). This indicated that IncM is involved in the ability of Chlamydia to inhibit host cell cytokinesis. The capacity of IncM to induce multinucleation in infected cells was shown to be conserved among its chlamydial homologues and appeared to require its two larger regions predicted to be exposed to the host cell cytosol. C. trachomatis-infected cells also displayed IncM-dependent defects in centrosome positioning, Golgi distribution around the inclusion, and morphology and stability of the inclusion. The altered morphology of inclusions containing IncM-deficient C. trachomatis was further affected by depolymerization of host cell microtubules. This was not observed after depolymerization of microfilaments, and inclusions containing wild-type C. trachomatis did not alter their morphology upon depolymerization of microtubules. Overall, these findings suggest that IncM may exert its effector function by acting directly or indirectly on host cell microtubules.


Subject(s)
Chlamydia Infections , Cytokinesis , Humans , Cytokinesis/physiology , Chlamydia trachomatis/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , HeLa Cells , Vacuoles/metabolism , Centrosome/metabolism , Chlamydia Infections/microbiology , Host-Pathogen Interactions
6.
Int J Biol Macromol ; 224: 55-67, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36252630

ABSTRACT

The cellulosome is an elaborate multi-enzyme structure secreted by many anaerobic microorganisms for the efficient degradation of lignocellulosic substrates. It is composed of multiple catalytic and non-catalytic components that are assembled through high-affinity protein-protein interactions between the enzyme-borne dockerin (Doc) modules and the repeated cohesin (Coh) modules present in primary scaffoldins. In some cellulosomes, primary scaffoldins can interact with adaptor and cell-anchoring scaffoldins to create structures of increasing complexity. The cellulosomal system of the ruminal bacterium, Ruminococcus flavefaciens, is one of the most intricate described to date. An unprecedent number of different Doc specificities results in an elaborate architecture, assembled exclusively through single-binding-mode type-III Coh-Doc interactions. However, a set of type-III Docs exhibits certain features associated with the classic dual-binding mode Coh-Doc interaction. Here, the structure of the adaptor scaffoldin-borne ScaH Doc in complex with the Coh from anchoring scaffoldin ScaE is described. This complex, unlike previously described type-III interactions in R. flavefaciens, was found to interact in a dual-binding mode. The key residues determining Coh recognition were also identified. This information was used to perform structure-informed protein engineering to change the electrostatic profile of the binding surface and to improve the affinity between the two modules. The results show that the nature of the residues in the ligand-binding surface plays a major role in Coh recognition and that Coh-Doc affinity can be manipulated through rational design, a key feature for the creation of designer cellulosomes or other affinity-based technologies using tailored Coh-Doc interactions.


Subject(s)
Bacterial Proteins , Cellulosomes , Amino Acid Sequence , Bacterial Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Cohesins
7.
ACS Chem Biol ; 17(7): 1901-1909, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35766974

ABSTRACT

Metal-dependent formate dehydrogenases are important enzymes due to their activity of CO2 reduction to formate. The tungsten-containing FdhAB formate dehydrogenase from Desulfovibrio vulgaris Hildenborough is a good example displaying high activity, simple composition, and a notable structural and catalytic robustness. Here, we report the first spectroscopic redox characterization of FdhAB metal centers by EPR. Titration with dithionite or formate leads to reduction of three [4Fe-4S]1+ clusters, and full reduction requires Ti(III)-citrate. The redox potentials of the four [4Fe-4S]1+ centers range between -250 and -530 mV. Two distinct WV signals were detected, WDV and WFV, which differ in only the g2-value. This difference can be explained by small variations in the twist angle of the two pyranopterins, as determined through DFT calculations of model compounds. The redox potential of WVI/V was determined to be -370 mV when reduced by dithionite and -340 mV when reduced by formate. The crystal structure of dithionite-reduced FdhAB was determined at high resolution (1.5 Å), revealing the same structural alterations as reported for the formate-reduced structure. These results corroborate a stable six-ligand W coordination in the catalytic intermediate WV state of FdhAB.


Subject(s)
Desulfovibrio vulgaris , Desulfovibrio , Catalysis , Desulfovibrio/metabolism , Desulfovibrio vulgaris/metabolism , Dithionite , Electron Spin Resonance Spectroscopy , Formate Dehydrogenases/chemistry , Formate Dehydrogenases/metabolism , Formates , Metals , Oxidation-Reduction
8.
Int J Mol Sci ; 24(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36613918

ABSTRACT

Metal-dependent formate dehydrogenases (Fdh) catalyze the reversible conversion of CO2 to formate, with unrivalled efficiency and selectivity. However, the key catalytic aspects of these enzymes remain unknown, preventing us from fully benefiting from their capabilities in terms of biotechnological applications. Here, we report a time-resolved characterization by X-ray crystallography of the Desulfovibrio vulgaris Hildenborough SeCys/W-Fdh during formate oxidation. The results allowed us to model five different intermediate structures and to chronologically map the changes occurring during enzyme reduction. Formate molecules were assigned for the first time to populate the catalytic pocket of a Fdh. Finally, the redox reversibility of DvFdhAB in crystals was confirmed by reduction and reoxidation structural studies.


Subject(s)
Formate Dehydrogenases , Metals , Formate Dehydrogenases/metabolism , Oxidation-Reduction , Catalysis , Formates/chemistry , Carbon Dioxide/chemistry
9.
J Med Chem ; 64(17): 13025-13037, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34415167

ABSTRACT

Human aldehyde oxidase (hAOX1) is mainly present in the liver and has an emerging role in drug metabolism, since it accepts a wide range of molecules as substrates and inhibitors. Herein, we employed an integrative approach by combining NMR, X-ray crystallography, and enzyme inhibition kinetics to understand the inhibition modes of three hAOX1 inhibitors-thioridazine, benzamidine, and raloxifene. These integrative data indicate that thioridazine is a noncompetitive inhibitor, while benzamidine presents a mixed type of inhibition. Additionally, we describe the first crystal structure of hAOX1 in complex with raloxifene. Raloxifene binds tightly at the entrance of the substrate tunnel, stabilizing the flexible entrance gates and elucidating an unusual substrate-dependent mechanism of inhibition with potential impact on drug-drug interactions. This study can be considered as a proof-of-concept for an efficient experimental screening of prospective substrates and inhibitors of hAOX1 relevant in drug discovery.


Subject(s)
Aldehyde Oxidase/antagonists & inhibitors , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Benzamidines/chemistry , Benzamidines/pharmacology , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation , Raloxifene Hydrochloride/chemistry , Selective Estrogen Receptor Modulators/chemistry , Thioridazine/chemistry , Thioridazine/pharmacology
10.
J Biol Chem ; 295(16): 5377-5389, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32144208

ABSTRACT

Aldehyde oxidases (AOXs) are a small group of enzymes belonging to the larger family of molybdo-flavoenzymes, along with the well-characterized xanthine oxidoreductase. The two major types of reactions that are catalyzed by AOXs are the hydroxylation of heterocycles and the oxidation of aldehydes to their corresponding carboxylic acids. Different animal species have different complements of AOX genes. The two extremes are represented in humans and rodents; whereas the human genome contains a single active gene (AOX1), those of rodents, such as mice, are endowed with four genes (Aox1-4), clustering on the same chromosome, each encoding a functionally distinct AOX enzyme. It still remains enigmatic why some species have numerous AOX enzymes, whereas others harbor only one functional enzyme. At present, little is known about the physiological relevance of AOX enzymes in humans and their additional forms in other mammals. These enzymes are expressed in the liver and play an important role in the metabolisms of drugs and other xenobiotics. In this review, we discuss the expression, tissue-specific roles, and substrate specificities of the different mammalian AOX enzymes and highlight insights into their physiological roles.


Subject(s)
Aldehyde Oxidase/metabolism , Evolution, Molecular , Liver/enzymology , Aldehyde Oxidase/chemistry , Aldehyde Oxidase/genetics , Animals , Humans , Substrate Specificity
11.
ACS Biomater Sci Eng ; 6(2): 1090-1101, 2020 02 10.
Article in English | MEDLINE | ID: mdl-33464857

ABSTRACT

CO-releasing molecules (CORMs) have been widely studied for their anti-inflammatory, antiapoptotic, and antiproliferative effects. CORM-3 is a water-soluble Ru-based metal carbonyl complex, which metallates serum proteins and readily releases CO in biological media. In this work, we evaluated the anti-inflammatory and wound-healing effects of gold nanoparticles-CORM-3 conjugates, AuNPs@PEG@BSA·Ru(CO)x, exploring its use as an efficient CO carrier. Our results suggest that the nanoformulation was capable of inducing a more pronounced cell effect, at the anti-inflammatory level and a faster tissue repair, probably derived from a rapid cell uptake of the nanoformulation that results in the increase of CO inside the cell.


Subject(s)
Coordination Complexes , Metal Nanoparticles , Organometallic Compounds , Carbon Monoxide , Gold
12.
Biochim Biophys Acta Gen Subj ; 1864(1): 129440, 2020 01.
Article in English | MEDLINE | ID: mdl-31536751

ABSTRACT

BACKGROUND: Half of human cancers harbour TP53 mutations that render p53 inactive as a tumor suppressor. As such, reactivation of mutant (mut)p53 through restoration of wild-type (wt)-like function represents one of the most promising therapeutic strategies in cancer treatment. Recently, we have reported the (S)-tryptophanol-derived oxazoloisoindolinone SLMP53-1 as a new reactivator of wt and mutp53 R280K with in vitro and in vivo p53-dependent antitumor activity. The present work aimed a mechanistic elucidation of mutp53 reactivation by SLMP53-1. METHODS AND RESULTS: By cellular thermal shift assay (CETSA), it is shown that SLMP53-1 induces wt and mutp53 R280K thermal stabilization, which is indicative of intermolecular interactions with these proteins. Accordingly, in silico studies of wt and mutp53 R280K DNA-binding domain with SLMP53-1 unveiled that the compound binds at the interface of the p53 homodimer with the DNA minor groove. Additionally, using yeast and p53-null tumor cells ectopically expressing distinct highly prevalent mutp53, the ability of SLMP53-1 to reactivate multiple mutp53 is evidenced. CONCLUSIONS: SLMP53-1 is a p53-activating agent with the ability to directly target wt and a set of hotspot mutp53. GENERAL SIGNIFICANCE: This work reinforces the encouraging application of SLMP53-1 in the personalized treatment of cancer patients harboring distinct p53 status.


Subject(s)
DNA-Binding Proteins/genetics , Isoindoles/pharmacology , Neoplasms/drug therapy , Oxazoles/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoindoles/chemistry , Mutation/drug effects , Neoplasms/genetics , Neoplasms/pathology , Oxazoles/chemistry , Protein Domains/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors
13.
FEBS Open Bio ; 9(5): 925-934, 2019 05.
Article in English | MEDLINE | ID: mdl-30985987

ABSTRACT

Human aldehyde oxidase (hAOX1) is a molybdenum enzyme with high toxicological importance, but its physiological role is still unknown. hAOX1 metabolizes different classes of xenobiotics and is one of the main drug-metabolizing enzymes in the liver, along with cytochrome P450. hAOX1 oxidizes and inactivates a large number of drug molecules and has been responsible for the failure of several phase I clinical trials. The interindividual variability of drug-metabolizing enzymes caused by single nucleotide polymorphisms (SNPs) is highly relevant in pharmaceutical treatments. In this study, we present the crystal structure of the inactive variant G1269R, revealing the first structure of a molybdenum cofactor (Moco)-free form of hAOX1. These data allowed to model, for the first time, the flexible Gate 1 that controls access to the active site. Furthermore, we inspected the thermostability of wild-type hAOX1 and hAOX1 with various SNPs (L438V, R1231H, G1269R or S1271L) by CD spectroscopy and ThermoFAD, revealing that amino acid exchanges close to the Moco site can impact protein stability up to 10 °C. These results correlated with biochemical and structural data and enhance our understanding of hAOX1 and the effect of SNPs in the gene encoding this enzyme in the human population. ENZYMES: Aldehyde oxidase (EC1.2.3.1); xanthine dehydrogenase (EC1.17.1.4); xanthine oxidase (EC1.1.3.2). DATABASES: Structural data are available in the Protein Data Bank under the accession number 6Q6Q.


Subject(s)
Aldehyde Oxidase/chemistry , Polymorphism, Single Nucleotide , Coenzymes , Crystallography, X-Ray , Humans , Metalloproteins , Models, Molecular , Molybdenum Cofactors , Pteridines
14.
Int J Mol Sci ; 19(4)2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29652801

ABSTRACT

The p53 tumor suppressor is widely found to be mutated in human cancer. This protein is regarded as a molecular hub regulating different cell responses, namely cell death. Compelling data have demonstrated that the impairment of p53 activity correlates with tumor development and maintenance. For these reasons, the reactivation of p53 function is regarded as a promising strategy to halt cancer. In the present work, the recombinant mutant p53R280K DNA binding domain (DBD) was produced for the first time, and its crystal structure was determined in the absence of DNA to a resolution of 2.0 Å. The solved structure contains four molecules in the asymmetric unit, four zinc(II) ions, and 336 water molecules. The structure was compared with the wild-type p53 DBD structure, isolated and in complex with DNA. These comparisons contributed to a deeper understanding of the mutant p53R280K structure, as well as the loss of DNA binding related to halted transcriptional activity. The structural information derived may also contribute to the rational design of mutant p53 reactivating molecules with potential application in cancer treatment.


Subject(s)
Arginine/genetics , DNA/metabolism , Lysine/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Crystallography, X-Ray , Humans , Hydrogen Bonding , Models, Molecular , Mutation , Protein Binding , Protein Structure, Secondary , Tumor Suppressor Protein p53/metabolism , Water , Zinc/chemistry
15.
Sci Rep ; 8(1): 5313, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593310

ABSTRACT

Gram-positive bacteria homeostasis and antibiotic resistance mechanisms are dependent on the intricate architecture of the cell wall, where amidated peptidoglycan plays an important role. The amidation reaction is carried out by the bi-enzymatic complex MurT-GatD, for which biochemical and structural information is very scarce. In this work, we report the first crystal structure of the glutamine amidotransferase member of this complex, GatD from Staphylococcus aureus, at 1.85 Å resolution. A glutamine molecule is found close to the active site funnel, hydrogen-bonded to the conserved R128. In vitro functional studies using 1H-NMR spectroscopy showed that S. aureus MurT-GatD complex has glutaminase activity even in the absence of lipid II, the MurT substrate. In addition, we produced R128A, C94A and H189A mutants, which were totally inactive for glutamine deamidation, revealing their essential role in substrate sequestration and catalytic reaction. GatD from S. aureus and other pathogenic bacteria share high identity to enzymes involved in cobalamin biosynthesis, which can be grouped in a new sub-family of glutamine amidotransferases. Given the ubiquitous presence of GatD, these results provide significant insights into the molecular basis of the so far undisclosed amidation mechanism, contributing to the development of alternative therapeutics to fight infections.


Subject(s)
Anthranilate Synthase/metabolism , Anthranilate Synthase/ultrastructure , Nitrogenous Group Transferases/metabolism , Nitrogenous Group Transferases/ultrastructure , Staphylococcus aureus/enzymology , Anti-Bacterial Agents/analysis , Bacterial Proteins/analysis , Carbon-Nitrogen Ligases , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Catalytic Domain , Cell Wall/chemistry , Glutamic Acid/metabolism , Glutamine/metabolism , Gram-Positive Bacteria , Multienzyme Complexes , Peptidoglycan/chemistry , Staphylococcal Infections , Staphylococcus aureus/metabolism
16.
Biochim Biophys Acta Bioenerg ; 1858(10): 865-872, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28801050

ABSTRACT

Arsenic is a widely distributed environmental toxin whose presence in drinking water poses a threat to >140 million people worldwide. The respiratory enzyme arsenite oxidase from various bacteria catalyses the oxidation of arsenite to arsenate and is being developed as a biosensor for arsenite. The arsenite oxidase from Rhizobium sp. str. NT-26 (a member of the Alphaproteobacteria) is a heterotetramer consisting of a large catalytic subunit (AioA), which contains a molybdenum centre and a 3Fe-4S cluster, and a small subunit (AioB) containing a Rieske 2Fe-2S cluster. Stopped-flow spectroscopy and isothermal titration calorimetry (ITC) have been used to better understand electron transfer through the redox-active centres of the enzyme, which is essential for biosensor development. Results show that oxidation of arsenite at the active site is extremely fast with a rate of >4000s-1 and reduction of the electron acceptor is rate-limiting. An AioB-F108A mutation results in increased activity with the artificial electron acceptor DCPIP and decreased activity with cytochrome c, which in the latter as demonstrated by ITC is not due to an effect on the protein-protein interaction but instead to an effect on electron transfer. These results provide further support that the AioB F108 is important in electron transfer between the Rieske subunit and cytochrome c and its absence in the arsenite oxidases from the Betaproteobacteria may explain the inability of these enzymes to use this electron acceptor.


Subject(s)
Cytochromes c/metabolism , Electron Transport/physiology , Oxidoreductases/metabolism , Arsenites/metabolism , Betaproteobacteria/metabolism , Catalysis , Catalytic Domain/physiology , Electrons , Molybdenum/metabolism , Oxidation-Reduction , Protein Interaction Maps/physiology , Protein Subunits/metabolism
17.
J Biol Chem ; 292(12): 4847-4860, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28179427

ABSTRACT

Deconstruction of cellulose, the most abundant plant cell wall polysaccharide, requires the cooperative activity of a large repertoire of microbial enzymes. Modular cellulases contain non-catalytic type A carbohydrate-binding modules (CBMs) that specifically bind to the crystalline regions of cellulose, thus promoting enzyme efficacy through proximity and targeting effects. Although type A CBMs play a critical role in cellulose recycling, their mechanism of action remains poorly understood. Here we produced a library of recombinant CBMs representative of the known diversity of type A modules. The binding properties of 40 CBMs, in fusion with an N-terminal GFP domain, revealed that type A CBMs possess the ability to recognize different crystalline forms of cellulose and chitin over a wide range of temperatures, pH levels, and ionic strengths. A Spirochaeta thermophila CBM64, in particular, displayed plasticity in its capacity to bind both crystalline and soluble carbohydrates under a wide range of extreme conditions. The structure of S. thermophila StCBM64C revealed an untwisted, flat, carbohydrate-binding interface comprising the side chains of four tryptophan residues in a co-planar linear arrangement. Significantly, two highly conserved asparagine side chains, each one located between two tryptophan residues, are critical to insoluble and soluble glucan recognition but not to bind xyloglucan. Thus, CBM64 compact structure and its extended and versatile ligand interacting platform illustrate how type A CBMs target their appended plant cell wall-degrading enzymes to a diversity of recalcitrant carbohydrates under a wide range of environmental conditions.


Subject(s)
Bacterial Proteins/metabolism , Cellulases/metabolism , Spirochaeta/metabolism , Bacterial Proteins/chemistry , Binding Sites , Carbohydrate Metabolism , Cell Wall/metabolism , Cellulases/chemistry , Cellulose/metabolism , Crystallography, X-Ray , Glucans/metabolism , Models, Molecular , Osmolar Concentration , Protein Binding , Protein Conformation , Spirochaeta/chemistry , Temperature , Xylans/metabolism
18.
Curr Opin Chem Biol ; 37: 39-47, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28126656

ABSTRACT

Aldehyde oxidases (AOXs) are molybdo-flavoenzymes characterized by broad substrate specificity, oxidizing aromatic/aliphatic aldehydes into the corresponding carboxylic acids and hydroxylating various heteroaromatic rings. Mammals are characterized by a complement of species-specific AOX isoenzymes, that varies from one in humans (AOX1) to four in rodents (AOX1, AOX2, AOX3 and AOX4). The physiological function of mammalian AOX isoenzymes is unknown, although human AOX1 is an emerging enzyme in phase-I drug metabolism. Indeed, the number of therapeutic molecules under development which act as AOX substrates is increasing. The recent crystallization and structure determination of human AOX1 as well as mouse AOX3 has brought new insights into the mechanisms underlying substrate/inhibitor binding as well as the catalytic activity of this class of enzymes.


Subject(s)
Aldehyde Oxidase/chemistry , Aldehyde Oxidase/metabolism , Mammals , Pharmaceutical Preparations/metabolism , Xenobiotics/metabolism , Aldehyde Oxidase/antagonists & inhibitors , Aldehyde Oxidase/genetics , Animals , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Polymorphism, Single Nucleotide
19.
Sci Rep ; 6: 38292, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27924829

ABSTRACT

During the course of evolution, the cellulosome, one of Nature's most intricate multi-enzyme complexes, has been continuously fine-tuned to efficiently deconstruct recalcitrant carbohydrates. To facilitate the uptake of released sugars, anaerobic bacteria use highly ordered protein-protein interactions to recruit these nanomachines to the cell surface. Dockerin modules located within a non-catalytic macromolecular scaffold, whose primary role is to assemble cellulosomal enzymatic subunits, bind cohesin modules of cell envelope proteins, thereby anchoring the cellulosome onto the bacterial cell. Here we have elucidated the unique molecular mechanisms used by anaerobic bacteria for cellulosome cellular attachment. The structure and biochemical analysis of five cohesin-dockerin complexes revealed that cell surface dockerins contain two cohesin-binding interfaces, which can present different or identical specificities. In contrast to the current static model, we propose that dockerins utilize multivalent modes of cohesin recognition to recruit cellulosomes to the cell surface, a mechanism that maximises substrate access while facilitating complex assembly.


Subject(s)
Bacterial Proteins/chemistry , Cell Cycle Proteins/chemistry , Cellulosomes/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Clostridiales/chemistry , Clostridium thermocellum/chemistry , Membrane Proteins/chemistry , Multienzyme Complexes/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , Cellulosomes/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cloning, Molecular , Clostridiales/metabolism , Clostridium thermocellum/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics , Cohesins
20.
ACS Chem Biol ; 11(10): 2923-2935, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27622978

ABSTRACT

The xanthine oxidase (XO) family comprises molybdenum-dependent enzymes that usually form homodimers (or dimers of heterodimers/trimers) organized in three domains that harbor two [2Fe-2S] clusters, one FAD, and a Mo cofactor. In this work, we crystallized an unusual member of the family, the periplasmic aldehyde oxidoreductase PaoABC from Escherichia coli. This is the first example of an E. coli protein containing a molybdopterin-cytosine-dinucleotide cofactor and is the only heterotrimer of the XO family so far structurally characterized. The crystal structure revealed the presence of an unexpected [4Fe-4S] cluster, anchored to an additional 40 residues subdomain. According to phylogenetic analysis, proteins containing this cluster are widely spread in many bacteria phyla, putatively through repeated gene transfer events. The active site of PaoABC is highly exposed to the surface with no aromatic residues and an arginine (PaoC-R440) making a direct interaction with PaoC-E692, which acts as a base catalyst. In order to understand the importance of R440, kinetic assays were carried out, and the crystal structure of the PaoC-R440H variant was also determined.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Escherichia coli/enzymology , Molybdenum/metabolism , Periplasm/enzymology , Xanthine Oxidase/metabolism , Aldehyde Dehydrogenase/chemistry , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...