Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biochim Biophys Acta ; 1834(12): 2600-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24063888

ABSTRACT

Hyperthermophilic enzymes are of industrial importance and interest, especially due to their denaturation kinetics at commercial sterilisation temperatures inside safety indicating time-temperature integrators (TTIs). The thermal stability and irreversible thermal inactivation of native extracellular Pyrococcus furiosus α-amylase were investigated using differential scanning calorimetry, circular dichroism and Fourier transform infrared spectroscopy. Denaturation of the amylase was irreversible above a Tm of approximately 106°C and could be described by a one-step irreversible model. The activation energy at 121°C was found to be 316kJ/mol. Using CD and FT-IR spectroscopy it was shown that folding and stability greatly increase with temperature. Under an isothermal holding temperature of 121°C, the structure of the PFA changes during denaturation from an α-helical structure, through a ß-sheet structure to an aggregated protein. Such data reinforces the use of P. furiosus α-amylase as a labile species in TTIs.


Subject(s)
Archaeal Proteins/chemistry , Hot Temperature , Models, Chemical , Protein Denaturation , Pyrococcus furiosus/enzymology , alpha-Amylases/chemistry , Enzyme Stability , Kinetics , Protein Structure, Secondary
2.
J Clin Invest ; 103(7): 999-1006, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10194472

ABSTRACT

The association between Z alpha1-antitrypsin deficiency and juvenile cirrhosis is well-recognized, and there is now convincing evidence that the hepatic inclusions are the result of entangled polymers of mutant Z alpha1-antitrypsin. Four percent of the northern European Caucasian population are heterozygotes for the Z variant, but even more common is S alpha1-antitrypsin, which is found in up to 28% of southern Europeans. The S variant is known to have an increased susceptibility to polymerization, although this is marginal compared with the more conformationally unstable Z variant. There has been speculation that the two may interact to produce cirrhosis, but this has never been demonstrated experimentally. This hypothesis was raised again by the observation reported here of a mixed heterozygote for Z alpha1-antitrypsin and another conformationally unstable variant (I alpha1-antitrypsin; 39Arg-->Cys) identified in a 34-year-old man with cirrhosis related to alpha1-antitrypsin deficiency. The conformational stability of the I variant has been characterized, and we have used fluorescence resonance energy transfer to demonstrate the formation of heteropolymers between S and Z alpha1-antitrypsin. Taken together, these results indicate that not only may mixed variants form heteropolymers, but that this can causally lead to the development of cirrhosis.


Subject(s)
Liver Cirrhosis/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/chemistry , Adult , Animals , Heterozygote , Humans , Liver Cirrhosis/pathology , Male , Microinjections , Models, Molecular , Mutation , Oocytes , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Messenger/genetics , White People , Xenopus
3.
J Mol Biol ; 296(1): 43-56, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10656817

ABSTRACT

Phage T7 DNA ligase seals nicked DNA substrates and is a representative member of the ATP-dependent class of DNA ligases. Although the catalytic mechanism of DNA ligases has been delineated, little is known about the nature of nick recognition by these enzymes. Here, we show that T7 ligase discriminates, at the nick-binding step, between nicks containing either a 5'-phosphate or a 5'-OH. T7 ligase binds preferentially to phosphorylated nicks and catalyses the sealing reaction. We also show using DNA footprinting studies, that T7 ligase binds asymmetrically to nicks as a monomer, with the protein interface covering between 12 and 14 bp of DNA. Based on molecular modelling studies we propose a structural model of the ligase-DNA complex consistent with these and other data. Using photo-crosslinking and site-directed mutagenesis we have identified two residues, K238 and K240, critical for the transadenylation and nick-sealing reactions. Sequence conservation and structural analysis supports the premise that these two lysine residues are critical for both nucleotide binding and DNA nick recognition. The implications of these results on the ligation mechanism are discussed.


Subject(s)
Bacteriophage T7/enzymology , DNA Damage/genetics , DNA Ligases/metabolism , DNA/genetics , DNA/metabolism , Amino Acid Sequence , Bacteriophage T7/genetics , Base Sequence , Binding Sites/radiation effects , Catalysis , Conserved Sequence/genetics , DNA/chemistry , DNA Footprinting , DNA Ligases/chemistry , DNA Ligases/genetics , DNA Ligases/isolation & purification , Lysine/genetics , Lysine/metabolism , Models, Molecular , Molecular Conformation , Mutation/genetics , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Phosphates/metabolism , Protein Binding/radiation effects , Substrate Specificity , Ultraviolet Rays
4.
J Mol Biol ; 305(4): 773-83, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11162091

ABSTRACT

Plasminogen activator inhibitor type 1 (PAI-1) is a member of the serine protease inhibitor (serpin) superfamily. Its highly mobile reactive-center loop (RCL) is thought to account for both the rapid inhibition of tissue-type plasminogen activator (t-PA), and the rapid and spontaneous transition of the unstable, active form of PAI-1 into a stable, inactive (latent) conformation (t(1/2) at 37 degrees C, 2.2 hours). We determined the amino acid residues responsible for the inherent instability of PAI-1, to assess whether these properties are independent and, consequently, whether the structural basis for inhibition and latency transition is different. For that purpose, a hypermutated PAI-1 library that is displayed on phage was pre-incubated for increasing periods (20 to 72 hours) at 37 degrees C, prior to a stringent selection for rapid t-PA binding. Accordingly, four rounds of phage-display selection resulted in the isolation of a stable PAI-1 variant (st-44: t(1/2) 450 hours) with 11 amino acid mutations. Backcrossing by DNA shuffling of this stable mutant with wt PAI-1 was performed to eliminate non-contributing mutations. It was shown that the combination of mutations at positions 50, 56, 61, 70, 94, 150, 222, 223, 264 and 331 increases the half-life of PAI-1 245-fold. Furthermore, within the limits of detection the stable mutants isolated are functionally indistinguishable from wild-type PAI-1 with respect to the rate of inhibition of t-PA, cleavage by t-PA, and binding to vitronectin. These stabilizing mutations constitute largely reversions to the stable "serpin consensus sequence" and are located in areas implicated in PAI-1 stability (e.g. the vitronectin-binding domain and the proximal hinge). Collectively, our data provide evidence that the structural requirements for PAI-1 loop insertion during latency transition and target proteinase inhibition can be separated.


Subject(s)
Mutagenesis/genetics , Peptide Library , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/metabolism , Tissue Plasminogen Activator/antagonists & inhibitors , Animals , Consensus Sequence , Half-Life , Humans , Kinetics , Mice , Models, Molecular , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/isolation & purification , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Thermodynamics , Tissue Plasminogen Activator/metabolism , Vitronectin/metabolism
5.
J Mol Biol ; 285(2): 703-12, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-10075524

ABSTRACT

The structural basis for the extreme discrimination achieved by malate dehydrogenases between a variety of closely related substrates encountered within the cell has been difficult to assess because of the lack of an appropriate catalytically competent structure of the enzyme. Here, we have determined the crystal structure of a ternary complex of porcine cytoplasmic malate dehydrogenase with the alternative substrate alpha-ketomalonate and the coenzyme analogue 1,4,5,6-tetrahydronicotinamide. Both subunits of the dimeric porcine heart, and from the prokaryotes Escherichia coli and Thermus flavus. However, large changes are noted around the active site, where a mobile loop now closes to bring key residues into contact with the substrate. This observation substantiates a postulated mechanism in which the enzyme achieves high levels of substrate discrimination through charge balancing in the active site. As the activated cofactor/substrate complex has a net negative charge, a positive counter-charge is provided by a conserved arginine in the active site loop. The enzyme must, however, also discriminate against smaller substrates, such as pyruvate. The structure shows in the closed (loop down) catalytically competent complex two arginine residues (91 and 97) are driven into close proximity. Without the complimentary, negative charge of the substrate side-chain of oxaloacetate or alpha-ketomalonate, charge repulsion would resist formation production of this catalytically productive conformation, hence minimising the effectiveness of pyruvate as a substrate. By this mechanism, malate dehydrogenase uses charge balancing to achieve fivefold orders of magnitude in discrimination between potential substrates.


Subject(s)
Malate Dehydrogenase/chemistry , Malonates/chemistry , NAD/analogs & derivatives , Protein Conformation , Amino Acid Sequence , Animals , Aspartic Acid , Binding Sites , Crystallography, X-Ray , Cytoplasm , Histidine , Malate Dehydrogenase/metabolism , Malonates/metabolism , Molecular Sequence Data , NAD/chemistry , NAD/metabolism , Protein Structure, Secondary , Substrate Specificity , Swine
6.
Protein Sci ; 7(3): 782-8, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9541411

ABSTRACT

The binding of heparin causes a conformational change in antithrombin to give an increased heparin binding affinity and activate the inhibition of thrombin and factor Xa. The areas of antithrombin involved in binding heparin and stabilizing the interaction in the high-affinity form have been partially resolved through the study of both recombinant and natural variants. The role of a section of the N-terminal segment of antithrombin, residues 22-46 (segment 22-46), in heparin binding was investigated using rapid kinetic analysis of the protein cleaved at residues 29-30 by limited proteolysis with thermolysin. The cleaved antithrombin had 5.5-fold lowered affinity for heparin pentasaccharide and 1.8-fold for full-length, high-affinity heparin. It was shown that, although the initial binding of heparin is slightly enhanced by the cleavage, it dissociates much faster from the cleaved form, giving rise to the overall decrease in heparin affinity. This implies that the segment constituting residues 22-46 in the N terminus of antithrombin hinders access to the binding site for heparin, hence the increased initial binding for the cleaved form, whereas, when heparin is bound, segment 22-46 is involved in the stabilization of the binding interaction, as indicated by the increased dissociation constant. When the heparin pentasaccharide is bound to antithrombin prior to incubation with thermolysin, it protects the N-terminal cleavage site, implying that segment 22-46 moves to interact with heparin in the conformational change and thus stabilizes the complex.


Subject(s)
Antithrombin III/metabolism , Heparin/metabolism , Humans , Kinetics , Models, Molecular , Oligosaccharides/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Thermolysin
7.
Protein Sci ; 9(7): 1274-81, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10933492

ABSTRACT

Members of the serpin family of serine proteinase inhibitors play important roles in the inflammatory, coagulation, fibrinolytic, and complement cascades. An inherent part of their function is the ability to undergo a structural rearrangement, the stressed (S) to relaxed (R) transition, in which an extra strand is inserted into the central A beta-sheet. In order for this transition to take place, the A sheet has to be unusually flexible. Malfunctions in this flexibility can lead to aberrant protein linkage, serpin inactivation, and diseases as diverse as cirrhosis, thrombosis, angioedema, emphysema, and dementia. The development of agents that control this conformational rearrangement requires a high resolution structure of an active serpin. We present here the topology of the archetypal serpin alpha1-antitrypsin to 2 A resolution. This structure allows us to define five cavities that are potential targets for rational drug design to develop agents that will prevent conformational transitions and ameliorate the associated disease.


Subject(s)
alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/metabolism , Crystallography, X-Ray , Drug Design , Models, Molecular , Mutation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha 1-Antitrypsin/genetics
8.
Chem Commun (Camb) ; (23): 2410-1, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-12239990

ABSTRACT

Flow linear dichroism is shown to be able to detect single base mismatches in a polymerase chain reaction (PCR) amplimers from exon 10 of the human beta-glucocerebrosidase gene (associated with Gaucher disease) over a kilobase long with no post PCR manipulation.


Subject(s)
Base Pair Mismatch , Gaucher Disease/genetics , Glucosylceramidase/genetics , Alleles , Exons/genetics , Humans , Polymerase Chain Reaction , Spectrum Analysis
9.
J Biol Chem ; 276(52): 49310-9, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11592970

ABSTRACT

Heat shock protein 47 (Hsp47) is a procollagen/collagen-specific molecular chaperone protein derived from the serpin family of proteins and essential for the early stages of collagen biosynthesis. In this paper, the results of an extensive biophysical analysis of mature recombinant mouse Hsp47 show the existence of both a structurally mesostable monomer with a 5-strand A-sheet and/or a hyperstable trimer; both states have biological activity. It is also demonstrated that Hsp47 is able to bind to a monomeric and partially folded conformation collagen mimic peptide (PPG)(10). Upon peptide binding Hsp47 has the capacity to induce the peptide backbone to fold into a polyproline type II conformation. Induction of this conformation results in (PPG)(10) peptides associating into higher order assemblies with increased stability compared with the monomeric peptide alone. These assemblies are similar to those observed by others (Go, N., and Suezaki, Y. (1973) Biopolymers 12, 1927-1930; Engel, J., Chen, H. T., Prockop, D. J., and Klump, H. (1977) Biopolymers 16, 601-622) when the peptide is dissolved at high concentration and are proposed to be long chains of peptides in a collagen-like configuration. Examination of the biophysical characteristics of both monomeric and trimeric Hsp47-(PPG)(10) complexes is also used to determine that the peptide-binding site does not reside in strand 4 position of sheet A, as observed for other serpins (Skinner, R., Chang, W. S. W., Jin, L., Pei, X., Huntington, J. A., Abrahams, J. P., Carrell, R. W., and Lomas, D. A. (1998) J. Mol. Biol. 283, 9-14), leaving earlier proposals of a binding site near helix A viable.


Subject(s)
Collagen/biosynthesis , Heat-Shock Proteins/metabolism , Protein Structure, Quaternary , Animals , Circular Dichroism , Electrophoresis , HSP47 Heat-Shock Proteins , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/isolation & purification , Humans , Mice , Models, Biological , Models, Molecular , Protein Binding , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Serpins/chemistry , Serpins/metabolism
10.
J Biol Chem ; 275(43): 33663-8, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10924508

ABSTRACT

alpha(1)-Antitrypsin is the most abundant circulating protease inhibitor and the archetype of the serine protease inhibitor or serpin superfamily. Members of this family may be inactivated by point mutations that favor transition to a polymeric conformation. This polymeric conformation underlies diseases as diverse as alpha(1)-antitrypsin deficiency-related cirrhosis, thrombosis, angio-edema, and dementia. The precise structural linkage within a polymer has been the subject of much debate with evidence for reactive loop insertion into beta-sheet A or C or as strand 7A. We have used site directed cysteine mutants and fluorescence resonance energy transfer (FRET) to measure a number of distances between monomeric units in polymeric alpha(1)-antitrypsin. We have then used a combinatorial approach to compare distances determined from FRET with distances obtained from 2.9 x 10(6) different possible orientations of the alpha(1)-antitrypsin polymer. The closest matches between experimental FRET measurements and theoretical structures show conclusively that polymers of alpha(1)-antitrypsin form by insertion of the reactive loop into beta-sheet A.


Subject(s)
alpha 1-Antitrypsin/chemistry , Fluorescence , Models, Molecular , Polymers/chemistry , Protein Structure, Secondary
11.
Protein Eng ; 10(1): 39-44, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9051732

ABSTRACT

This paper describes the testing of a homology model of Plasmodium falciparum lactate dehydrogenase (pfLDH) by protein engineering. The model had been validated in structural terms. It suggests explanations of the unusual properties of pfLDH (compared with all other LDHs). These unusual features are a lack of substrate inhibition, high activity with the synthetic coenzyme 3-acetylpyridine adenine dinucleotide (APAD+) and changes in residues at previously conserved positions. pfLDH shows several amino acid insertions and deletions in an alignment with protein sequences from all other known LDHs. The most notable is a five amino acid insertion into the active-site loop. In addition, a conserved serine at position 163 is replaced by leucine. The results showed that when the unique pfLDH structural features were engineered into Bacillus stearothermophilus lactate dehydrogenase, the thermophilic enzyme acquired the properties previously uniquely associated with the malarial enzyme. We conclude that the homology model of the malarial enzyme is adequate for the prediction of successful redesigns and, in the regions tested, is accurate.


Subject(s)
L-Lactate Dehydrogenase/genetics , Plasmodium falciparum/enzymology , Amino Acid Sequence , Animals , Binding Sites/physiology , Drug Design , Geobacillus stearothermophilus/enzymology , Geobacillus stearothermophilus/genetics , Kinetics , L-Lactate Dehydrogenase/pharmacokinetics , L-Lactate Dehydrogenase/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , NAD/analogs & derivatives , NAD/metabolism , Plasmodium falciparum/genetics , Protein Engineering , Sequence Homology, Amino Acid , Structure-Activity Relationship
12.
J Biol Chem ; 275(12): 8733-41, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722716

ABSTRACT

Antithrombin requires heparin for efficient inhibition of the final two proteinases of the blood coagulation cascade, factor Xa and thrombin. Antithrombin binds heparin via a specific pentasaccharide domain in a two-step mechanism whereby initial weak binding is followed by a conformational change and subsequent tight binding. The goal of this study is to investigate the role of a reducing-end extension in the binding of the longer oligosaccharides that contain the cognate pentasaccharide sequence. We determined the antithrombin binding properties of a synthetic heptasaccharide containing the natural pentasaccharide sequence (DEFGH) and an additional reducing-end disaccharide (DEFGHG'H'). Binding at low ionic strength is unaffected by the disaccharide addition, but at ionic strengths >/=0.2 the mode of heptasaccharide binding changes resulting in a 2-fold increase in affinity due to a decrease in the off-rate caused by a greater nonionic contribution to binding. Molecular modeling of possible binding modes for the heptasaccharide at high ionic strength indicates a possible shift in position of the pentasaccharide domain to occupy the extended heparin-binding site. This conclusion supports the likely presence of a range of sequences that can bind to and activate antithrombin in the natural heparan sulfates that line the vascular endothelium.


Subject(s)
Antithrombins/metabolism , Heparin/metabolism , Oligosaccharides/metabolism , Antithrombins/drug effects , Binding Sites , Carbohydrate Sequence , Heparin/chemistry , Heparin/pharmacology , Iduronic Acid , Kinetics , Models, Chemical , Models, Molecular , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Protein Conformation
13.
J Biol Chem ; 274(14): 9548-55, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10092640

ABSTRACT

The mutation in the Z deficiency variant of alpha1-antitrypsin perturbs the structure of the protein to allow a unique intermolecular linkage. These loop-sheet polymers are retained within the endoplasmic reticulum of hepatocytes to form inclusions that are associated with neonatal hepatitis, juvenile cirrhosis, and hepatocellular carcinoma. The process of polymer formation has been investigated here by intrinsic tryptophan fluorescence, fluorescence polarization, circular dichroic spectra and extrinsic fluorescence with 8-anilino-1-naphthalenesulfonic acid and tetramethylrhodamine-5-iodoacetamide. These biophysical techniques have demonstrated that alpha1-antitrypsin polymerization is a two-stage process and have allowed the calculation of rates for both of these steps. The initial fast phase is unimolecular and likely to represent temperature-induced protein unfolding, while the slow phase is bimolecular and associated with loop-sheet interaction and polymer formation. The naturally occurring Z, S, and I variants and recombinant site-directed reactive loop and shutter domain mutants of alpha1-antitrypsin were used to demonstrate the close association between protein stability and rate of alpha1-antitrypsin polymerization. Taken together, these data allow us to propose a kinetic mechanism for alpha1-antitrypsin polymer formation that involves the generation of an unstable intermediate, which can form polymers or generate latent protein.


Subject(s)
Polymers/metabolism , alpha 1-Antitrypsin/metabolism , Circular Dichroism , Fluorescence Polarization , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Molecular Mimicry , Point Mutation , Protein Conformation , Protein Folding , alpha 1-Antitrypsin/genetics
14.
J Biol Chem ; 276(12): 9115-22, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11102455

ABSTRACT

The activity of the serine proteinase inhibitor (serpin) plasminogen activator inhibitor-1 (PAI-1) is controlled by the intramolecular incorporation of the reactive loop into beta-sheet A with the generation of an inactive latent species. Other members of the serpin superfamily can be pathologically inactivated by intermolecular linkage between the reactive loop of one molecule and beta-sheet A of a second to form chains of polymers associated with diverse diseases. It has long been believed that PAI-1 is unique among active serpins in that it does not form polymers. We show here that recombinant native and latent PAI-1 spontaneously form polymers in vitro at low pH although with distinctly different electrophoretic patterns of polymerization. The polymers of both the native and latent species differ from the typical loop-A-sheet polymers of other serpins in that they readily dissociate back to their original monomeric form. The findings with PAI-1 are compatible with different mechanisms of linkage, each involving beta-strand addition of the reactive loop to s7A in native PAI-1 and to s1C in latent PAI-1. Glycosylated native and latent PAI-1 can also form polymers under similar conditions, which may be of in vivo importance in the low pH environment of the platelet.


Subject(s)
Biopolymers/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Amino Acid Sequence , Biopolymers/chemistry , Chromatography, Gel , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/isolation & purification , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
15.
Proteins ; 44(3): 329-35, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11455606

ABSTRACT

An homology model of protochlorophyllide reductase (POR) from Synechocystis sp. was constructed on a template from the tyrosine-dependent oxidoreductase family. The model showed characteristics appropriate to a globular, soluble protein and was used to generate a structure of the ternary complex of POR, nicotinamide adenine dinucleotide phosphate (NADPH), and protochlorophyllide. The POR ternary model was validated by mutagenesis experiments involving predicted coenzyme-binding residues and by chemical modification experiments. A core tryptophan residue was shown to be responsible for much of the protein's fluorescence. Both quenching of this residue by coenzyme and fluorescence resonance energy transfer (FRET) from the protein to the coenzyme allowed the binding constant of NADPH to be determined. Replacement of this residue by Tyr gave an active mutant with approximately halved fluorescence and a negligible FRET signal, consistent with the role of this residue in energy transfer to the NADPH at the active site and with the model. The mechanism of the enzyme is discussed in the context of the model and semiempirical molecular orbital calculations.


Subject(s)
Cyanobacteria/enzymology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/chemistry , Amino Acid Sequence , Binding Sites , Fluorescence , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , NADP/metabolism , Oxidoreductases/genetics , Sequence Homology, Amino Acid
16.
Proteins ; 29(2): 228-39, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9329087

ABSTRACT

Current methods for reengineering enzyme substrate specificities rely heavily on the use of static x-ray crystallographic models. In this article we detail the use of a molecular mechanics approach for suggesting regions of Bacillus stearothermophilus L-lactate dehydrogenase (EC 1.1.1.27) involved in substrate specificity, and hence areas of interest for protein engineers. The approach combines molecular dynamics with energy minimization (MD/EM) to search the conformational space available to a 15-A sphere of the ternary complex centered on the catalytic histidine. The search is carried out by calculating a 30-ps dynamics trajectory at 300 K and minimizing structures at 1-ps intervals. The protocol has been performed on 14 systems containing different combinations of substrate and mutant/wt LDH. In order to discover which interactions are important in defining substrate specificity, eight conformational parameters representing substrate-active site interactions were measured in each of the 420 minimized structures. These parameters were then compared to the measured catalytic activity of the protein-substrate combinations. These comparisons show that arginine 109 orientation is a major determining factor in LDH specificity. Using this methodology it is possible to estimate the catalytic activity of proteins of varied sequence by computer simulation before synthesis.


Subject(s)
Geobacillus stearothermophilus/enzymology , L-Lactate Dehydrogenase/metabolism , Catalysis , Kinetics , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/genetics , Mutagenesis , Protein Conformation , Substrate Specificity
17.
Tissue Antigens ; 63(4): 345-54, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15009806

ABSTRACT

Human leukocyte antigen (HLA) A2 is one of the most immunodominant HLA antigens. Through a process of light-chain variable domain (VL) shuffling, we analyzed the VL domains' role in anti-HLA-A2/A28-binding site diversity. This was achieved by combining a VH3-30-encoded HLA-A2/A28-specific heavy-chain variable domain with 10(4) non-immune VL domains. Twelve HLA-A2/A28-specific antibodies were subsequently identified. VL gene analysis demonstrated an absence of Vlambda domains and that all have VkappaI-encoded light chains. The affinities correlated with the VkappaI gene present, with the seven highest affinity antibodies using Vkappa domains encoded by the O18 gene segment. A 300-fold difference in affinity was observed between the 12 antibodies, and homology modeling demonstrated a correlation between electrostatic surface potential of the antigen-binding site and affinity for HLA. Overlap between the T-cell receptor-binding site and that of the antibodies was indicated by inhibition of cytotoxic T-lymphocyte killing of peptide-pulsed target cells. A model of antibody binding to HLA-A2 suggested contact with both alpha helices of the HLA molecule, such that the antigen-binding site spans the peptide-binding groove. These data increase the understanding of antibody recognition of HLA and may facilitate the production of clonotypic antibodies with peptide-specific binding.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Epitopes/immunology , HLA-A2 Antigen/immunology , Immunoglobulin Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Affinity/genetics , Binding Sites, Antibody , Cloning, Molecular , Cytotoxicity Tests, Immunologic , Gene Library , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Protein Structure, Tertiary , Structural Homology, Protein
18.
Proc Natl Acad Sci U S A ; 97(1): 67-72, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10618372

ABSTRACT

The serpins are a family of proteinase inhibitors that play a central role in the control of proteolytic cascades. Their inhibitory mechanism depends on the intramolecular insertion of the reactive loop into beta-sheet A after cleavage by the target proteinase. Point mutations within the protein can allow aberrant conformational transitions characterized by beta-strand exchange between the reactive loop of one molecule and beta-sheet A of another. These loop-sheet polymers result in diseases as varied as cirrhosis, emphysema, angio-oedema, and thrombosis, and we recently have shown that they underlie an early-onset dementia. We report here the biochemical characteristics and crystal structure of a naturally occurring variant (Leu-55-Pro) of the plasma serpin alpha(1)-antichymotrypsin trapped as an inactive intermediate. The structure demonstrates a serpin configuration with partial insertion of the reactive loop into beta-sheet A. The lower part of the sheet is filled by the last turn of F-helix and the loop that links it to s3A. This conformation matches that of proposed intermediates on the pathway to complex and polymer formation in the serpins. In particular, this intermediate, along with the latent and polymerized conformations, explains the loss of activity of plasma alpha(1)-antichymotrypsin associated with chronic obstructive pulmonary disease in patients with the Leu-55-Pro mutation.


Subject(s)
alpha 1-Antichymotrypsin/chemistry , Chromatography, Affinity , Humans , Lung Diseases, Obstructive/blood , Lung Diseases, Obstructive/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Plasmids , Protein Conformation , Protein Structure, Secondary , X-Ray Diffraction , alpha 1-Antichymotrypsin/blood , alpha 1-Antichymotrypsin/genetics
19.
Cell ; 97(6): 805-15, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10380931

ABSTRACT

The alpha subunit of the endocytotic AP2 adaptor complex contains a 30 kDa "appendage" domain, which is joined to the rest of the protein via a flexible linker. The 1.9 A resolution crystal structure of this domain reveals a single binding site for its ligands, which include amphiphysin, Eps15, and epsin. This domain when overexpressed in COS7 fibroblasts is shown to inhibit transferrin uptake, whereas mutants in which interactions with its binding partners are abolished do not. DPF/W motifs present in appendage domain-binding partners are shown to play a crucial role in their interactions with the domain. A single site for binding multiple ligands would allow for temporal and spatial regulation in the recruitment of components of the endocytic machinery.


Subject(s)
Membrane Proteins/chemistry , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Crystallography, X-Ray , Endocytosis , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Conformation
20.
Blood ; 92(8): 2696-706, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9763552

ABSTRACT

The inherent variability of conformational diseases is demonstrated by two families with different mutations of the same conserved aminoacid in antithrombin. Threonine 85 underlies the opening of the main beta-sheet of the molecule and its replacement, by the polar lysine, in antithrombin Wobble, resulted in a plasma deficiency of antithrombin with an uncharacteristically severe onset of thrombosis at 10 years of age, whereas the replacement of the same residue by a nonpolar methionine, antithrombin Wibble, gave near-normal levels of plasma antithrombin and more typical adult thromboembolic disease. Isolated antithrombin Wibble had a decreased thermal stability (Tm 56.2, normal 57.6 degreesC) but was fully stabilized by the heparin pentasaccharide (Tm 71.8, normal 71.0 degreesC), indicating that the prime abnormality is a laxity in the transition of the main sheet of the molecule from the 5- to 6-stranded form, as was confirmed by the ready conversion of antithrombin Wibble to the 6-stranded latent form on incubation. That this transition can occur in vivo was shown by the finding of nearly 10% of the proband's plasma antithrombin in the latent form and also, surprisingly, of small but definitive amounts of latent antithrombin in normal plasma. The latent transition will be predictably accelerated not only by gross mutations, as with antithrombin Wobble, to give severe episodic thrombosis, but also by milder mutations, as with antithrombin Wibble, to trigger thrombosis in the presence of other predisposing factors, including the conformational stress imposed by the raised body temperatures of fevers. Both antithrombin variants had an exceptional (25-fold) increase in heparin affinity and this, together with an increased inhibitory activity against factor Xa, provides evidence of the direct linkage of A-sheet opening to the conformational basis of heparin binding and activation.


Subject(s)
Amino Acid Substitution , Antithrombin III Deficiency/genetics , Antithrombins/genetics , Mutation, Missense , Point Mutation , Protein Conformation , Antithrombin III Deficiency/blood , Antithrombin Proteins , Antithrombins/chemistry , Child , DNA, Complementary/metabolism , Factor V/genetics , Female , Heparin/metabolism , Hot Temperature , Humans , Male , Middle Aged , Oligosaccharides/metabolism , Protein Binding , Protein Denaturation , Structure-Activity Relationship , Threonine
SELECTION OF CITATIONS
SEARCH DETAIL