Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
ACS Catal ; 12(18): 11444-11455, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36158901

ABSTRACT

A 2-keto-3-deoxygluconate aldolase from the hyperthermophile Sulfolobus solfataricus catalyzes the nonstereoselective aldol reaction of pyruvate and d-glyceraldehyde to produce 2-keto-3-deoxygluconate (d-KDGlc) and 2-keto-3-deoxy-d-galactonate (d-KDGal). Previous investigations into curing the stereochemical promiscuity of this hyperstable aldolase used high-resolution structures of the aldolase bound to d-KDGlc or d-KDGal to identify critical amino acids involved in substrate binding for mutation. This structure-guided approach enabled mutant variants to be created that could stereoselectively catalyze the aldol reaction of pyruvate and natural d-glyceraldehyde to selectively afford d-KDGlc or d-KDGal. Here we describe the creation of two further mutants of this Sulfolobus aldolase that can be used to catalyze aldol reactions between pyruvate and non-natural l-glyceraldehyde to enable the diastereoselective synthesis of l-KDGlc and l-KDGal. High-resolution crystal structures of all four variant aldolases have been determined (both unliganded and liganded), including Variant 1 with d-KDGlc, Variant 2 with pyruvate, Variant 3 with l-KDGlc, and Variant 4 with l-KDGal. These structures have enabled us to rationalize the observed changes in diastereoselectivities in these variant-catalyzed aldol reactions at a molecular level. Interestingly, the active site of Variant 4 was found to be sufficiently flexible to enable catalytically important amino acids to be replaced while still retaining sufficient enzymic activity to enable production of l-KDGal.

2.
Front Immunol ; 13: 892234, 2022.
Article in English | MEDLINE | ID: mdl-35693766

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen that is able to thwart an effective host immune response by producing a range of immune evasion molecules, including S. aureus binder of IgG (Sbi) which interacts directly with the central complement component C3, its fragments and associated regulators. Recently we reported the first structure of a disulfide-linked human C3d17C dimer and highlighted its potential role in modulating B-cell activation. Here we present an X-ray crystal structure of a disulfide-linked human C3d17C dimer, which undergoes a structurally stabilising N-terminal 3D domain swap when in complex with Sbi. These structural studies, in combination with circular dichroism and fluorescence spectroscopic analyses, reveal the mechanism underpinning this unique helix swap event and could explain the origins of a previously discovered N-terminally truncated C3dg dimer isolated from rat serum. Overall, our study unveils a novel staphylococcal complement evasion mechanism which enables the pathogen to harness the ability of dimeric C3d to modulate B-cell activation.


Subject(s)
Bacterial Proteins , Staphylococcus aureus , Animals , Carrier Proteins/metabolism , Disulfides/metabolism , Rats , Staphylococcus/metabolism
3.
Front Immunol ; 12: 714055, 2021.
Article in English | MEDLINE | ID: mdl-34434196

ABSTRACT

Cleavage of C3 to C3a and C3b plays a central role in the generation of complement-mediated defences. Although the thioester-mediated surface deposition of C3b has been well-studied, fluid phase dimers of C3 fragments remain largely unexplored. Here we show C3 cleavage results in the spontaneous formation of C3b dimers and present the first X-ray crystal structure of a disulphide-linked human C3d dimer. Binding studies reveal these dimers are capable of crosslinking complement receptor 2 and preliminary cell-based analyses suggest they could modulate B cell activation to influence tolerogenic pathways. Altogether, insights into the physiologically-relevant functions of C3d(g) dimers gained from our findings will pave the way to enhancing our understanding surrounding the importance of complement in the fluid phase and could inform the design of novel therapies for immune system disorders in the future.


Subject(s)
Complement C3d/chemistry , Models, Molecular , Protein Multimerization , Complement C3/chemistry , Complement C3/immunology , Complement C3d/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Proteolysis , Recombinant Proteins/chemistry , Structure-Activity Relationship
4.
Eur J Med Chem ; 158: 25-33, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30199703

ABSTRACT

The trans-sialidase protein expressed by Trypanosoma cruzi is an important enzyme in the life cycle of this human pathogenic parasite and is considered a promising target for the development of new drug treatments against Chagas' disease. Here we describe α-amino phosphonates as a novel class of inhibitor of T. cruzi trans-sialidase. Molecular modelling studies were initially used to predict the active-site binding affinities for a series of amino phosphonates, which were subsequently synthesised and their IC50s determined in vitro. The measured inhibitory activities show some correlation with the predictions from molecular modelling, with 1-napthyl derivatives found to be the most potent inhibitors having IC50s in the low micromolar range. Interestingly, kinetic analysis of the mode of inhibition demonstrated that the α-aminophosphonates tested here operate in a non-competitive manner.


Subject(s)
Chagas Disease/drug therapy , Glycoproteins/antagonists & inhibitors , Neuraminidase/antagonists & inhibitors , Organophosphonates/chemistry , Organophosphonates/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/enzymology , Amination , Chagas Disease/parasitology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycoproteins/chemistry , Glycoproteins/metabolism , Humans , Molecular Docking Simulation , Neuraminidase/chemistry , Neuraminidase/metabolism , Trypanosoma cruzi/drug effects
5.
J Biol Chem ; 293(24): 9335-9344, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29724824

ABSTRACT

Kidney- and brain-expressed protein (KIBRA), a multifunctional scaffold protein with around 20 known binding partners, is involved in memory and cognition, organ size control via the Hippo pathway, cell polarity, and membrane trafficking. KIBRA includes tandem N-terminal WW domains, a C2 domain, and motifs for binding atypical PKC and PDZ domains. A naturally occurring human KIBRA variant involving residue changes at positions 734 (Met-to-Ile) and 735 (Ser-to-Ala) within the C2 domain affects cognitive performance. We have elucidated 3D structures and calcium- and phosphoinositide-binding properties of human KIBRA C2 domain. Both WT and variant C2 adopt a canonical type I topology C2 domain fold. Neither Ca2+ nor any other metal ion was bound to WT or variant KIBRA C2 in crystal structures, and Ca2+ titration produced no significant reproducible changes in NMR spectra. NMR and X-ray diffraction data indicate that KIBRA C2 binds phosphoinositides via an atypical site involving ß-strands 5, 2, 1, and 8. Molecular dynamics simulations indicate that KIBRA C2 interacts with membranes via primary and secondary sites on the same domain face as the experimentally identified phosphoinositide-binding site. Our results indicate that KIBRA C2 domain association with membranes is calcium-independent and involves distinctive C2 domain-membrane relative orientations.


Subject(s)
Calcium/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphatidylinositols/metabolism , Phosphoproteins/metabolism , C2 Domains , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation
6.
Biochemistry ; 57(26): 3797-3806, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29812914

ABSTRACT

The thermoacidophilic archaea Picrophilus torridus and Sulfolobus solfataricus catabolize glucose via a nonphosphorylative Entner-Doudoroff pathway and a branched Entner-Doudoroff pathway, respectively. Key enzymes for these Entner-Doudoroff pathways are the aldolases, 2-keto-3-deoxygluconate aldolase (KDG-aldolase) and 2-keto-3-deoxy-6-phosphogluconate aldolase [KD(P)G-aldolase]. KDG-aldolase from P. torridus (Pt-KDG-aldolase) is highly specific for the nonphosphorylated substrate, 2-keto-3-deoxygluconate (KDG), whereas KD(P)G-aldolase from S. solfataricus [Ss-KD(P)G-aldolase] is an enzyme that catalyzes the cleavage of both KDG and 2-keto-3-deoxy-6-phosphogluconate (KDPG), with a preference for KDPG. The structural basis for the high specificity of Pt-KDG-aldolase for KDG as compared to the more promiscuous Ss-KD(P)G-aldolase has not been analyzed before. In this work, we report the elucidation of the structure of Ss-KD(P)G-aldolase in complex with KDPG at 2.35 Å and that of KDG-aldolase from P. torridus at 2.50 Å resolution. By superimposition of the active sites of the two enzymes, and subsequent site-directed mutagenesis studies, a network of four amino acids, namely, Arg106, Tyr132, Arg237, and Ser241, was identified in Ss-KD(P)G-aldolase that interact with the negatively charged phosphate group of KDPG, thereby increasing the affinity of the enzyme for KDPG. This KDPG-binding network is absent in Pt-KDG-aldolase, which explains the low catalytic efficiency of KDPG cleavage.


Subject(s)
Aldehyde-Lyases/chemistry , Archaeal Proteins/chemistry , Gluconates/chemistry , Sulfolobus solfataricus/enzymology , Thermoplasmales/enzymology , Models, Molecular , Protein Domains , Structure-Activity Relationship
7.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 3): 179-186, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29497023

ABSTRACT

Pyruvate decarboxylase (PDC; EC 4.1.1.1) is a key enzyme in homofermentative metabolism where ethanol is the major product. PDCs are thiamine pyrophosphate- and Mg2+ ion-dependent enzymes that catalyse the non-oxidative decarboxylation of pyruvate to acetaldehyde and carbon dioxide. As this enzyme class is rare in bacteria, current knowledge of bacterial PDCs is extremely limited. One approach to further the understanding of bacterial PDCs is to exploit the diversity provided by evolution. Ancestral sequence reconstruction (ASR) is a method of computational molecular evolution to infer extinct ancestral protein sequences, which can then be synthesized and experimentally characterized. Through ASR a novel PDC was generated, designated ANC27, that shares only 78% amino-acid sequence identity with its closest extant homologue (Komagataeibacter medellinensis PDC, GenBank accession No. WP_014105323.1), yet is fully functional. Crystals of this PDC diffracted to 3.5 Šresolution. The data were merged in space group P3221, with unit-cell parameters a = b = 108.33, c = 322.65 Å, and contained two dimers (two tetramer halves) in the asymmetric unit. The structure was solved by molecular replacement using PDB entry 2wvg as a model, and the final R values were Rwork = 0.246 (0.3671 in the highest resolution bin) and Rfree = 0.319 (0.4482 in the highest resolution bin). Comparison with extant bacterial PDCs supports the previously observed correlation between decreased tetramer interface area (and number of interactions) and decreased thermostability.


Subject(s)
Acetobacteraceae/enzymology , Pyruvate Decarboxylase/chemistry , Acetobacteraceae/classification , Amino Acid Sequence , Catalytic Domain , Crystallization , Crystallography, X-Ray , Models, Molecular , Protein Conformation
8.
Biochim Biophys Acta Proteins Proteom ; 1866(2): 292-306, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29155107

ABSTRACT

Hydrolysis of arabinoxylan (AX) by glycoside hydrolase family 10 (GH10) xylanases produces xylo- and arabinoxylo-oligosaccharides ((A)XOS) which have shown prebiotic effects. The thermostable GH10 xylanase RmXyn10A has shown great potential to produce (A)XOS. In this study, the structure of RmXyn10A was investigated, the catalytic module by homology modelling and site-directed mutagenesis and the arrangement of its five domains by small-angle X-ray scattering (SAXS). Substrate specificity was explored in silico by manual docking and molecular dynamic simulations. It has been shown in the literature that the glycone subsites of GH10 xylanases are well conserved and our results suggest that RmXyn10A is no exception. The aglycone subsites are less investigated, and the modelled structure of RmXyn10A suggests that loop ß6α6 in the aglycone part of the active site contains a non-conserved α-helix, which blocks the otherwise conserved space of subsite +2. This structural feature has only been observed for one other GH10 xylanase. In RmXyn10A, docking revealed two alternative binding regions, one on either side of the α-helix. However, only one was able to accommodate arabinose-substitutions and the mutation study suggests that the same region is responsible for binding XOS. Several non-conserved structural features are most likely to be responsible for providing affinity for arabinose-substitutions in subsites +1 and +2. The SAXS rigid model of the modular arrangement of RmXyn10A displays the catalytic module close to the cell-anchoring domain while the carbohydrate binding modules are further away, likely explaining the observed lack of contribution of the CBMs to activity.


Subject(s)
Bacterial Proteins/genetics , Endo-1,4-beta Xylanases/chemistry , Rhodothermus/enzymology , Bacterial Proteins/chemistry , Endo-1,4-beta Xylanases/genetics , Protein Domains , Protein Structure, Secondary , Rhodothermus/genetics
9.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 9): 700-6, 2016 09.
Article in English | MEDLINE | ID: mdl-27599861

ABSTRACT

Pyruvate decarboxylase (PDC; EC 4.1.1.1) is a thiamine pyrophosphate- and Mg(2+) ion-dependent enzyme that catalyses the non-oxidative decarboxylation of pyruvate to acetaldehyde and carbon dioxide. It is rare in bacteria, but is a key enzyme in homofermentative metabolism, where ethanol is the major product. Here, the previously unreported crystal structure of the bacterial pyruvate decarboxylase from Zymobacter palmae is presented. The crystals were shown to diffract to 2.15 Šresolution. They belonged to space group P21, with unit-cell parameters a = 204.56, b = 177.39, c = 244.55 Šand Rr.i.m. = 0.175 (0.714 in the highest resolution bin). The structure was solved by molecular replacement using PDB entry 2vbi as a model and the final R values were Rwork = 0.186 (0.271 in the highest resolution bin) and Rfree = 0.220 (0.300 in the highest resolution bin). Each of the six tetramers is a dimer of dimers, with each monomer sharing its thiamine pyrophosphate across the dimer interface, and some contain ethylene glycol mimicking the substrate pyruvate in the active site. Comparison with other bacterial PDCs shows a correlation of higher thermostability with greater tetramer interface area and number of interactions.


Subject(s)
Bacterial Proteins/chemistry , Halomonadaceae/chemistry , Magnesium/chemistry , Pyruvate Decarboxylase/chemistry , Pyruvic Acid/chemistry , Thiamine Pyrophosphate/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cations, Divalent , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Ethylene Glycol/chemistry , Gene Expression , Halomonadaceae/enzymology , Kinetics , Magnesium/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Pyruvate Decarboxylase/genetics , Pyruvate Decarboxylase/metabolism , Pyruvic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Thiamine Pyrophosphate/metabolism
10.
Protein Sci ; 25(11): 2045-2053, 2016 11.
Article in English | MEDLINE | ID: mdl-27571338

ABSTRACT

Acetylating aldehyde dehydrogenases (AcAldDH) catalyse the acetylation of Coenzyme-A (CoA), or in reverse generate acetaldehyde from Acetyl-CoA using NADH as a co-factor. This article reports the expression, purification, enzyme assay, and X-ray crystal structures of an AcAldDH from Geobacillus thermoglucosidasius (GtAcAldDH) to 2.1Å and in complex with CoA and NAD+ to 4.0Å. In the structure, the AcAldDH forms a close-knit dimer, similar to that seen in other Alcohol Dehydrogenase (ADH) structures. In GtAcAldDH, these dimers associate via their N-termini to form weakly interacting tetramers. This mode of tetrameric association is also seen in an unpublished AcAldDH deposited in the PDB, but is in contrast to all other ADH structures, (including the one other published AcAldDH found in a bacterial microcompartment), in which the dimers bury a large surface area including the C-termini. This novel mode of association sequesters the active sites and potentially reactive acyl-enzyme intermediates in the center of the tetramer. In other respects, the structure is very similar to the other AcAldDH, binding the cofactors in a corresponding fashion. This similarity enabled the identification of a shortened substrate cavity in G. thermoglucosidasius AcAldDH, explaining the limitations on the length of substrate accepted by the enzyme.


Subject(s)
Aldehyde Oxidoreductases/chemistry , Bacterial Proteins/chemistry , Coenzyme A/chemistry , Geobacillus stearothermophilus/enzymology , NAD/chemistry , Crystallography, X-Ray , Protein Domains
11.
Biochem J ; 463(3): 405-12, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25088564

ABSTRACT

The four-component polypeptides of the 2-oxoacid dehydrogenase complex from the thermophilic archaeon Thermoplasma acidophilum assemble to give an active multienzyme complex possessing activity with the branched-chain 2-oxoacids derived from leucine, isoleucine and valine, and with pyruvate. The dihydrolipoyl acyl-transferase (E2) core of the complex is composed of identical trimer-forming units that assemble into a novel 42-mer structure comprising octahedral and icosahedral geometric aspects. From our previously determined structure of this catalytic core, the inter-trimer interactions involve a tyrosine residue near the C-terminus secured in a hydrophobic pocket of an adjacent trimer like a ball-and-socket joint. In the present study, we have deleted the five C-terminal amino acids of the E2 polypeptide (IIYEI) and shown by equilibrium centrifugation that it now only assembles into a trimeric enzyme. This was confirmed by SAXS analysis, although this technique showed the presence of approximately 20% hexamers. The crystal structure of the trimeric truncated E2 core has been determined and shown to be virtually identical with the ones observed in the 42-mer, demonstrating that removal of the C-terminal anchor does not significantly affect the individual monomer or trimer structures. The truncated E2 is still able to bind both 2-oxoacid decarboxylase (E1) and dihydrolipoamide dehydrogenase (E3) components to give an active complex with catalytic activity similar to the native multienzyme complex. This is the first report of an active mini-complex for this enzyme, and raises the question of why all 2-oxoacid dehydrogenase complexes assemble into such large structures.


Subject(s)
Archaeal Proteins/chemistry , Multienzyme Complexes/chemistry , Oxidoreductases/chemistry , Thermoplasma/enzymology , Archaeal Proteins/genetics , Crystallography, X-Ray , Dihydrolipoamide Dehydrogenase/chemistry , Enzyme Stability , Hot Temperature , Kinetics , Multienzyme Complexes/genetics , Oxidoreductases/genetics , Protein Conformation , Scattering, Small Angle
12.
Proteins ; 82(10): 2657-70, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24948467

ABSTRACT

Citrate synthase (CS) catalyses the entry of carbon into the citric acid cycle and is highly-conserved structurally across the tree of life. Crystal structures of dimeric CSs are known in both "open" and "closed" forms, which differ by a substantial domain motion that closes the substrate-binding clefts. We explore both the static rigidity and the dynamic flexibility of CS structures from mesophilic and extremophilic organisms from all three evolutionary domains. The computational expense of this wide-ranging exploration is kept to a minimum by the use of rigidity analysis and rapid all-atom simulations of flexible motion, combining geometric simulation and elastic network modeling. CS structures from thermophiles display increased structural rigidity compared with the mesophilic enzyme. A CS structure from a psychrophile, stabilized by strong ionic interactions, appears to display likewise increased rigidity in conventional rigidity analysis; however, a novel modified analysis, taking into account the weakening of the hydrophobic effect at low temperatures, shows a more appropriate decreased rigidity. These rigidity variations do not, however, affect the character of the flexible dynamics, which are well conserved across all the structures studied. Simulation trajectories not only duplicate the crystallographically observed symmetric open-to-closed transitions, but also identify motions describing a previously unidentified antisymmetric functional motion. This antisymmetric motion would not be directly observed in crystallography but is revealed as an intrinsic property of the CS structure by modeling of flexible motion. This suggests that the functional motion closing the binding clefts in CS may be independent rather than symmetric and cooperative.


Subject(s)
Bacterial Proteins/chemistry , Citrate (si)-Synthase/chemistry , Models, Molecular , Animals , Arthrobacter/enzymology , Arthrobacter/growth & development , Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Citrate (si)-Synthase/metabolism , Databases, Protein , Enzyme Stability , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Dynamics Simulation , Protein Conformation , Pyrobaculum/enzymology , Pyrobaculum/growth & development , Pyrococcus furiosus/enzymology , Pyrococcus furiosus/growth & development , Sulfolobus solfataricus/enzymology , Sulfolobus solfataricus/growth & development , Sus scrofa , Thermoplasma/enzymology , Thermoplasma/growth & development , Thermus thermophilus/enzymology , Thermus thermophilus/growth & development
13.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 5): 1366-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24816105

ABSTRACT

Geobacillus thermoglucosidasius is a thermophilic bacterium that is able to ferment both C6 and C5 sugars to produce ethanol. During growth on hemicellulose biomass, an intracellular ß-xylosidase catalyses the hydrolysis of xylo-oligosaccharides to the monosaccharide xylose, which can then enter the pathways of central metabolism. The gene encoding a G. thermoglucosidasius ß-xylosidase belonging to CAZy glycoside hydrolase family GH52 has been cloned and expressed in Escherichia coli. The recombinant enzyme has been characterized and a high-resolution (1.7 Å) crystal structure has been determined, resulting in the first reported structure of a GH52 family member. A lower resolution (2.6 Å) structure of the enzyme-substrate complex shows the positioning of the xylobiose substrate to be consistent with the proposed retaining mechanism of the family; additionally, the deep cleft of the active-site pocket, plus the proximity of the neighbouring subunit, afford an explanation for the lack of catalytic activity towards the polymer xylan. Whilst the fold of the G. thermoglucosidasius ß-xylosidase is completely different from xylosidases in other CAZy families, the enzyme surprisingly shares structural similarities with other glycoside hydrolases, despite having no more than 13% sequence identity.


Subject(s)
Geobacillus/enzymology , Xylosidases/chemistry , Xylosidases/metabolism , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Disaccharides/chemistry , Disaccharides/metabolism , Escherichia coli/genetics , Models, Molecular , Protein Conformation , Xylosidases/genetics
14.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 10): 2104-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24100328

ABSTRACT

Bifunctional alcohol/aldehyde dehydrogenase (ADHE) enzymes are found within many fermentative microorganisms. They catalyse the conversion of an acyl-coenzyme A to an alcohol via an aldehyde intermediate; this is coupled to the oxidation of two NADH molecules to maintain the NAD(+) pool during fermentative metabolism. The structure of the alcohol dehydrogenase (ADH) domain of an ADHE protein from the ethanol-producing thermophile Geobacillus thermoglucosidasius has been determined to 2.5 Šresolution. This is the first structure to be reported for such a domain. In silico modelling has been carried out to generate a homology model of the aldehyde dehydrogenase domain, and this was subsequently docked with the ADH-domain structure to model the structure of the complete ADHE protein. This model suggests, for the first time, a structural mechanism for the formation of the large multimeric assemblies or `spirosomes' that are observed for this ADHE protein and which have previously been reported for ADHEs from other organisms.


Subject(s)
Alcohol Dehydrogenase/chemistry , Biofuels/microbiology , Ethanol , Geobacillus/enzymology , Models, Molecular , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/isolation & purification , Amino Acid Sequence , Crystallography, X-Ray , Fermentation , Geobacillus/genetics , Geobacillus/growth & development , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics
15.
Biochem J ; 449(2): 415-25, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23116157

ABSTRACT

Lipoylation, the covalent attachment of lipoic acid to 2-oxoacid dehydrogenase multi-enzyme complexes, is essential for metabolism in aerobic bacteria and eukarya. In Escherichia coli, lipoylation is catalysed by LplA (lipoate protein ligase) or by LipA (lipoic acid synthetase) and LipB [lipoyl(octanoyl) transferase] combined. Whereas bacterial and eukaryotic LplAs comprise a single two-domain protein, archaeal LplA function typically involves two proteins, LplA-N and LplA-C. In the thermophilic archaeon Thermoplasma acidophilum, LplA-N and LplA-C are encoded by overlapping genes in inverted orientation (lpla-c is upstream of lpla-n). The T. acidophilum LplA-N structure is known, but the LplA-C structure is unknown and LplA-C's role in lipoylation is unclear. In the present study, we have determined the structures of the substrate-free LplA-N-LplA-C complex and E2lipD (dihydrolipoyl acyltransferase lipoyl domain) that is lipoylated by LplA-N-LplA-C, and carried out biochemical analyses of this archaeal lipoylation system. Our data reveal the following: (i) LplA-C is disordered but folds upon association with LplA-N; (ii) LplA-C induces a conformational change in LplA-N involving substantial shortening of a loop that could repress catalytic activity of isolated LplA-N; (iii) the adenylate-binding region of LplA-N-LplA-C includes two helices rather than the purely loop structure of varying order observed in other LplA structures; (iv) LplAN-LplA-C and E2lipD do not interact in the absence of substrate; (v) LplA-N-LplA-C undergoes a conformational change (the details of which are currently undetermined) during lipoylation; and (vi) LplA-N-LplA-C can utilize octanoic acid as well as lipoic acid as substrate. The elucidated functional inter-dependence of LplA-N and LplA-C is consistent with their evolutionary co-retention in archaeal genomes.


Subject(s)
Archaeal Proteins/metabolism , Peptide Synthases/metabolism , Protein Processing, Post-Translational , Thermoplasma/enzymology , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Binding Sites , Crystallography, X-Ray , Dihydrolipoyllysine-Residue Acetyltransferase/chemistry , Dihydrolipoyllysine-Residue Acetyltransferase/genetics , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Lipoylation , Magnetic Resonance Spectroscopy , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Peptide Synthases/chemistry , Peptide Synthases/genetics , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Thioctic Acid/chemistry , Thioctic Acid/metabolism
16.
FEBS J ; 279(5): 713-23, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188654

ABSTRACT

The dihydrolipoyl acyl-transferase (E2) enzyme forms the structural and catalytic core of the tripartite 2-oxoacid dehydrogenase multienzyme complexes of the central metabolic pathways. Although this family of multienzyme complexes shares a common architecture, their E2 cores form homo-trimers that, depending on the source, further associate into either octahedral (24-mer) or icosahedral (60-mer) assemblies, as predicted by the principles of quasi-equivalence. In the crystal structure of the E2 core from Thermoplasma acidophilum, a thermophilic archaeon, the homo-trimers assemble into a unique 42-mer oblate spheroid. Analytical equilibrium centrifugation and small-angle X-ray scattering analyses confirm that this catalytically active 1.08 MDa assembly exists as a single species in solution, forming a hollow spheroid with a maximum diameter of 220 Å. In this paper we show that a monodisperse macromolecular assembly, built from identical subunits in non-identical environments, forms an irregular protein shell via non-equivalent interactions. This unusually irregular protein shell, combining cubic and dodecahedral geometrical elements, expands on the concept of quasi-equivalence as a basis for understanding macromolecular assemblies by showing that cubic point group symmetry is not a physical requirement in multienzyme assembly. These results extend our basic knowledge of protein assembly and greatly expand the number of possibilities to manipulate self-assembling biological complexes to be utilized in innovative nanotechnology applications.


Subject(s)
Archaeal Proteins/metabolism , Multienzyme Complexes/metabolism , Thermoplasma/enzymology , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Protein Conformation
17.
Extremophiles ; 15(3): 327-36, 2011 May.
Article in English | MEDLINE | ID: mdl-21424517

ABSTRACT

Using citrate synthase from the hyperthermophile Pyrococcus furiosus (PfCS) as our test molecule, we show through guanidine hydrochloride-induced unfolding that the dimer separates into folded, but inactive, monomers before individual subunit unfolding takes place. Given that forces across the dimer interface are vital for thermostability, a robust computational method was derived that uses the University of Houston Brownian Dynamics (UHBD) program to calculate both the hydrophobic and electrostatic contribution to the dimerisation energy at 100°C. The results from computational and experimental determination of the lowered stability of interface mutants were correlated, being both of the same order of magnitude and placing the mutant proteins in the same order of stability. This computational method, optimised for hyperthermophilic molecules and tested in the laboratory, after further testing on other examples, could be of widespread use in the prediction of thermostabilising mutations in other oligomeric proteins for which dissociation is the first step in unfolding.


Subject(s)
Archaeal Proteins/chemistry , Citrate (si)-Synthase/chemistry , Pyrococcus furiosus/enzymology , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Computer Simulation , Enzyme Stability , Guanidine/chemistry , Hot Temperature , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutation , Protein Folding , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits , Pyrococcus furiosus/genetics , Spectrometry, Fluorescence , Static Electricity , Structure-Activity Relationship , Surface Properties , Ultracentrifugation
18.
J Am Chem Soc ; 132(33): 11753-8, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20684556

ABSTRACT

2-Keto-3-deoxygluconate aldolase from the hyperthermophile Sulfolobus solfataricus is a highly thermostable type I aldolase that can catalyze carbon-carbon bond formation using nonphosphorylated substrates. However, it exhibits poor diastereocontrol in many of its aldol reactions, including the reaction of its natural substrates, pyruvate and D-glyceraldehyde, which afford a 55:45 mixture of D-2-keto-3-deoxygluconate (D-KDGlu) and D-2-keto-3-deoxy-galactonate (D-KDGal). We have employed detailed X-ray crystallographic structural information of this aldolase bound to these diastereoisomeric aldol products to selectively target specific amino acids for mutation for the rapid creation of stereochemically complementary mutants that catalyze either (Re)- or (Si)-facial selective aldol reactions to afford either D-KDGlu or D-KDGal with good levels of diastereocontrol.


Subject(s)
Aldehyde-Lyases/chemistry , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Biocatalysis , Crystallography, X-Ray , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Stereoisomerism , Sulfolobus solfataricus/enzymology
19.
J Mol Biol ; 384(5): 1400-7, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-18976662

ABSTRACT

Fab 35PA83 is an antibody fragment of non-human primate origin that neutralizes the anthrax lethal toxin. Human antibodies are usually preferred when clinical use is envisioned, even though their framework regions (FR) may carry mutations introduced during affinity maturation. These hypermutations can be immunogenic and therefore FR that are encoded by human germline genes, encountered in IgMs and thus part of the "self" proteins, are preferable. Accordingly, the proportion of FR residues in 35PA83 that were encoded by human V and J germline genes, i.e. the germinality index (GI) of 35PA83, was increased in a multistep cumulative approach. In a first step, the FR1 and FR4 residues of 35PA83 were changed simultaneously into their counterparts coded by 35PA83's closest human germline genes, without prior modelling. The resulting derivative of 35PA83 had the same affinity as its parental Fab. In a second step, the 3D structures of this first 35PA83 derivative, carrying the same type of residue changes but in the FR2 and FR3 regions, were modelled in silico from sequences. Some of the changes in FR2 or FR3 modified the predicted peptide backbone. The changes that did not seem to alter the structure were introduced simultaneously in the Fab by an in vitro method and resulted in a loss of reactivity, which could however be fully restored by a single point mutation. The final 35PA83 derivative had a GI higher than that of a fully human Fab, which had neutralization properties similar to 35PA83 and which was used as a benchmark in this study.


Subject(s)
Antibodies/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Computational Biology , Germ Cells/immunology , Primates/immunology , Protein Engineering , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibody Affinity/immunology , Cell Line , Humans , Immunoglobulin Variable Region/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Point Mutation/genetics , Protein Structure, Secondary , Sequence Alignment
20.
Res Microbiol ; 157(6): 569-74, 2006.
Article in English | MEDLINE | ID: mdl-16844348

ABSTRACT

We characterised pneumococcal neuraminidase A (NanA) by determining key amino acids required for the enzymatic activity of the protein. Single replacement of two residues, hypothesised to be important for the catalytic activity of neuraminidases, resulted in total loss of activity (E647 with Q or Y752 with F). The mutation of R663 to H caused substantial reduction in the catalytic ability of the enzyme. The inactive neuraminidases thus produced were protective immunogens against pneumococcal pneumonia in mice.


Subject(s)
Amino Acids/genetics , Bacterial Proteins/metabolism , Neuraminidase/metabolism , Streptococcus pneumoniae/enzymology , Amino Acid Sequence , Amino Acid Substitution , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Catalysis , Enzyme Activation , Female , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuraminidase/genetics , Neuraminidase/immunology , Pneumococcal Vaccines , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/prevention & control , Sequence Homology, Amino Acid , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...