Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Biometals ; 36(3): 683-702, 2023 06.
Article in English | MEDLINE | ID: mdl-36418809

ABSTRACT

Iron is an essential element for various lifeforms but is largely insoluble due to the oxygenation of Earth's atmosphere and oceans during the Proterozoic era. Metazoans evolved iron transport glycoproteins, like transferrin (Tf) and lactoferrin (Lf), to keep iron in a non-toxic, usable form, while maintaining a low free iron concentration in the body that is unable to sustain bacterial growth. To survive on the mucosal surfaces of the human respiratory tract where it exclusively resides, the Gram-negative bacterial pathogen Moraxella catarrhalis utilizes surface receptors for acquiring iron directly from human Tf and Lf. The receptors are comprised of a surface lipoprotein to capture iron-loaded Tf or Lf and deliver it to a TonB-dependent transporter (TBDT) for removal of iron and transport across the outer membrane. The subsequent transport of iron into the cell is normally mediated by a periplasmic iron-binding protein and inner membrane transport complex, which has yet to be determined for Moraxella catarrhalis. We identified two potential periplasm to cytoplasm transport systems and performed structural and functional studies with the periplasmic binding proteins (FbpA and AfeA) to evaluate their role. Growth studies with strains deleted in the fbpA or afeA gene demonstrated that FbpA, but not AfeA, was required for growth on human Tf or Lf. The crystal structure of FbpA with bound iron in the open conformation was obtained, identifying three tyrosine ligands that were required for growth on Tf or Lf. Computational modeling of the YfeA homologue, AfeA, revealed conserved residues involved in metal binding.


Subject(s)
Iron , Lactoferrin , Moraxella catarrhalis , Transferrin , Humans , Bacterial Proteins/metabolism , Iron/metabolism , Lactoferrin/metabolism , Transferrin/metabolism
2.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 10): 363-370, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36189720

ABSTRACT

Succinyl-CoA synthetase (SCS) catalyzes a three-step reaction in the citric acid cycle with succinyl-phosphate proposed as a catalytic intermediate. However, there are no structural data to show the binding of succinyl-phosphate to SCS. Recently, the catalytic mechanism underlying acetyl-CoA production by ATP-citrate lyase (ACLY) has been debated. The enzyme belongs to the family of acyl-CoA synthetases (nucleoside diphosphate-forming) for which SCS is the prototype. It was postulated that the amino-terminal portion catalyzes the full reaction and the carboxy-terminal portion plays only an allosteric role. This interpretation was based on the partial loss of the catalytic activity of ACLY when Glu599 was mutated to Gln or Ala, and on the interpretation that the phospho-citryl-CoA intermediate was trapped in the 2.85 Šresolution structure from cryogenic electron microscopy (cryo-EM). To better resolve the structure of the intermediate bound to the E599Q mutant, the equivalent mutation, E105αQ, was made in human GTP-specific SCS. The structure of the E105αQ mutant shows succinyl-phosphate bound to the enzyme at 1.58 Šresolution when the mutant, after phosphorylation in solution by Mg2+-ATP, was crystallized in the presence of magnesium ions, succinate and desulfo-CoA. The E105αQ mutant is still active but has a specific activity that is 120-fold less than that of the wild-type enzyme, with apparent Michaelis constants for succinate and CoA that are 50-fold and 11-fold higher, respectively. Based on this high-resolution structure, the cryo-EM maps of the E599Q ACLY complex reported previously should have revealed the binding of citryl-phosphate and CoA and not phospho-citryl-CoA.


Subject(s)
ATP Citrate (pro-S)-Lyase , Succinate-CoA Ligases , ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Acetyl Coenzyme A , Acyl Coenzyme A , Adenosine Triphosphate/metabolism , Crystallography, X-Ray , Diphosphates , Guanosine Triphosphate/metabolism , Humans , Magnesium , Multienzyme Complexes , Nucleosides , Oxo-Acid-Lyases , Succinate-CoA Ligases/chemistry , Succinates , Succinic Acid/metabolism
3.
Acta Crystallogr D Struct Biol ; 77(Pt 3): 357-368, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33645539

ABSTRACT

Succinyl-CoA synthetase (SCS) catalyzes a reversible reaction that is the only substrate-level phosphorylation in the citric acid cycle. One of the essential steps for the transfer of the phosphoryl group involves the movement of the phosphohistidine loop between active site I, where CoA, succinate and phosphate bind, and active site II, where the nucleotide binds. Here, the first crystal structure of SCS revealing the conformation of the phosphohistidine loop in site II of the porcine GTP-specific enzyme is presented. The phosphoryl transfer bridges a distance of 29 Šbetween the binding sites for phosphohistidine in site I and site II, so these crystal structures support the proposed mechanism of catalysis by SCS. In addition, a second succinate-binding site was discovered at the interface between the α- and ß-subunits of SCS, and another magnesium ion was found that interacts with the side chains of Glu141ß and Glu204ß via water-mediated interactions. These glutamate residues interact with the active-site histidine residue when it is bound in site II.


Subject(s)
Histidine/analogs & derivatives , Succinate-CoA Ligases/chemistry , Animals , Binding Sites , Biocatalysis , Crystallization , Crystallography, X-Ray , Glutamic Acid/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Histidine/chemistry , Magnesium/chemistry , Models, Molecular , Protein Conformation , Succinic Acid/chemistry , Swine
4.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 7): 302-308, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32627745

ABSTRACT

Succinyl-CoA synthetase (SCS) catalyzes the only substrate-level phosphorylation step in the tricarboxylic acid cycle. Human GTP-specific SCS (GTPSCS), an αß-heterodimer, was produced in Escherichia coli. The purified protein crystallized from a solution containing tartrate, CoA and magnesium chloride, and a crystal diffracted to 1.52 Šresolution. Tartryl-CoA was discovered to be bound to GTPSCS. The CoA portion lies in the amino-terminal domain of the α-subunit and the tartryl end extends towards the catalytic histidine residue. The terminal carboxylate binds to the phosphate-binding site of GTPSCS.


Subject(s)
Coenzyme A/chemistry , Guanosine Triphosphate/chemistry , Succinate-CoA Ligases/chemistry , Tartrates/chemistry , Amino Acid Sequence , Binding Sites , Coenzyme A/metabolism , Crystallography, X-Ray , Dimerization , Escherichia coli/metabolism , Histidine/chemistry , Humans , Magnesium Chloride , Models, Molecular , Phosphates/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Recombinant Proteins , Succinate-CoA Ligases/metabolism
5.
Protein Sci ; 28(10): 1840-1849, 2019 10.
Article in English | MEDLINE | ID: mdl-31411782

ABSTRACT

ATP-citrate lyase (ACLY) catalyzes production of acetyl-CoA and oxaloacetate from CoA and citrate using ATP. In humans, this cytoplasmic enzyme connects energy metabolism from carbohydrates to the production of lipids. In certain bacteria, ACLY is used to fix carbon in the reductive tricarboxylic acid cycle. The carboxy(C)-terminal portion of ACLY shows sequence similarity to citrate synthase of the tricarboxylic acid cycle. To investigate the roles of residues of ACLY equivalent to active site residues of citrate synthase, these residues in ACLY from Chlorobium limicola were mutated, and the proteins were investigated using kinetics assays and biophysical techniques. To obtain the crystal structure of the C-terminal portion of ACLY, full-length C. limicola ACLY was cleaved, first non-specifically with chymotrypsin and subsequently with Tobacco Etch Virus protease. Crystals of the C-terminal portion diffracted to high resolution, providing structures that show the positions of active site residues and how ACLY tetramerizes.


Subject(s)
ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Binding Sites , Biocatalysis , Chlorobium/enzymology , Crystallography, X-Ray , Models, Molecular , Mutagenesis, Site-Directed
6.
Acta Crystallogr D Struct Biol ; 75(Pt 7): 647-659, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31282474

ABSTRACT

Succinyl-CoA synthetase (SCS) catalyzes the only step of the tricarboxylic acid cycle that leads to substrate-level phosphorylation. Some forms of SCS are specific for ADP/ATP or for GDP/GTP, while others can bind all of these nucleotides, generally with different affinities. The theory of `gatekeeper' residues has been proposed to explain the nucleotide-specificity. Gatekeeper residues lie outside the binding site and create specific electrostatic interactions with incoming nucleotides to determine whether the nucleotides can enter the binding site. To test this theory, the crystal structure of the nucleotide-binding domain in complex with Mg2+-ADP was determined, as well as the structures of four proteins with single mutations, K46ßE, K114ßD, V113ßL and L227ßF, and one with two mutations, K46ßE/K114ßD. The crystal structures show that the enzyme is specific for ADP/ATP because of interactions between the nucleotide and the binding site. Nucleotide-specificity is provided by hydrogen-bonding interactions between the adenine base and Gln20ß, Gly111ß and Val113ß. The O atom of the side chain of Gln20ß interacts with N6 of ADP, while the side-chain N atom interacts with the carbonyl O atom of Gly111ß. It is the different conformations of the backbone at Gln20ß, of the side chain of Gln20ß and of the linker that make the enzyme ATP-specific. This linker connects the two subdomains of the ATP-grasp fold and interacts differently with adenine and guanine bases. The mutant proteins have similar conformations, although the L227ßF mutant shows structural changes that disrupt the binding site for the magnesium ion. Although the K46ßE/K114ßD double mutant of Blastocystis hominis SCS binds GTP better than ATP according to kinetic assays, only the complex with Mg2+-ADP was obtained.


Subject(s)
Adenosine Triphosphate/metabolism , Blastocystis hominis/enzymology , Models, Molecular , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/genetics , Succinate-CoA Ligases/metabolism , Binding Sites , Crystallography, X-Ray/methods , Escherichia coli/genetics , Fluorometry/methods , Hydrogen Bonding , Kinetics , Mutation , Protein Binding , Protein Domains
7.
Acta Crystallogr D Struct Biol ; 73(Pt 8): 660-671, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28777081

ABSTRACT

Hydroxycitrate from the fruit of Garcinia cambogia [i.e. (2S,3S)-2-hydroxycitrate] is the best-known inhibitor of ATP-citrate lyase. Well diffracting crystals showing how the inhibitor binds to human ATP-citrate lyase were grown by modifying the protein. The protein was modified by introducing cleavage sites for Tobacco etch virus protease on either side of a disordered linker. The protein crystallized consisted of residues 2-425-ENLYFQ and S-488-810 of human ATP-citrate lyase. (2S,3S)-2-Hydroxycitrate binds in the same orientation as citrate, but the citrate-binding domain (residues 248-421) adopts a different orientation with respect to the rest of the protein (residues 4-247, 490-746 and 748-809) from that previously seen. For the first time, electron density was evident for the loop that contains His760, which is phosphorylated as part of the catalytic mechanism. The pro-S carboxylate of (2S,3S)-2-hydroxycitrate is available to accept a phosphoryl group from His760. However, when co-crystals were grown with ATP and magnesium ions as well as either the inhibitor or citrate, Mg2+-ADP was bound and His760 was phosphorylated. The phosphoryl group was not transferred to the organic acid. This led to the interpretation that the active site is trapped in an open conformation. The strategy of designing cleavage sites to remove disordered residues could be useful in determining the crystal structures of other proteins.


Subject(s)
ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Citrates/metabolism , Binding Sites , Catalytic Domain , Citrates/chemistry , Crystallography, X-Ray , Fruit/chemistry , Fruit/metabolism , Garcinia cambogia/chemistry , Garcinia cambogia/metabolism , Humans , Magnesium/chemistry , Magnesium/metabolism , Molecular Docking Simulation , Protein Binding , Protein Domains
8.
Acta Crystallogr D Struct Biol ; 72(Pt 8): 912-21, 2016 08.
Article in English | MEDLINE | ID: mdl-27487822

ABSTRACT

Succinyl-CoA synthetase catalyzes the only step in the citric acid cycle that provides substrate-level phosphorylation. Although the binding sites for the substrates CoA, phosphate, and the nucleotides ADP and ATP or GDP and GTP have been identified, the binding site for succinate has not. To determine this binding site, pig GTP-specific succinyl-CoA synthetase was crystallized in the presence of succinate, magnesium ions and CoA, and the structure of the complex was determined by X-ray crystallography to 2.2 Šresolution. Succinate binds in the carboxy-terminal domain of the ß-subunit. The succinate-binding site is near both the active-site histidine residue that is phosphorylated in the reaction and the free thiol of CoA. The carboxy-terminal domain rearranges when succinate binds, burying this active site. However, succinate is not in position for transfer of the phosphoryl group from phosphohistidine. Here, it is proposed that when the active-site histidine residue has been phosphorylated by GTP, the phosphohistidine displaces phosphate and triggers the movement of the carboxylate of succinate into position to be phosphorylated. The structure shows why succinyl-CoA synthetase is specific for succinate and does not react appreciably with citrate nor with the other C4-dicarboxylic acids of the citric acid cycle, fumarate and oxaloacetate, but shows some activity with L-malate.


Subject(s)
Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Succinic Acid/metabolism , Animals , Binding Sites , Catalytic Domain , Coenzyme A/metabolism , Crystallography, X-Ray , Guanosine Triphosphate/metabolism , Magnesium/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Swine
9.
PLoS One ; 7(11): e49225, 2012.
Article in English | MEDLINE | ID: mdl-23152878

ABSTRACT

Tetraloop-receptor interactions are prevalent structural units in RNAs, and include the GAAA/11-nt and GNRA-minor groove interactions. In this study, we have compiled a set of 78 nonredundant loop-helix interactions from X-ray crystal structures, and examined them for the extent of their sequence and structural variation. Of the 78 interactions in the set, only four were classical GAAA/11-nt motifs, while over half (48) were GNRA-minor groove interactions. The GNRA-minor groove interactions were not a homogeneous set, but were divided into five subclasses. The most predominant subclass is characterized by two triple base pair interactions in the minor groove, flanked by two ribose zipper contacts. This geometry may be considered the "standard" GNRA-minor groove interaction, while the other four subclasses are alternative ways to form interfaces between a minor groove and tetraloop. The remaining 26 structures in the set of 78 have loops interacting with mostly idiosyncratic receptors. Among the entire set, a number of sequence-structure correlations can be identified, which may be used as initial hypotheses in predicting three-dimensional structures from primary sequences. Conversely, other sequence patterns are not predictive; for example, GAAA loop sequences and GG/CC receptors bind to each other with three distinct geometries. Finally, we observe an example of structural evolution in group II introns, in which loop-receptor motifs are substituted for each other while maintaining the larger three-dimensional geometry. Overall, the study gives a more complete view of RNA loop-helix interactions that exist in nature.


Subject(s)
Nucleic Acid Conformation , RNA/chemistry , RNA/metabolism , Base Sequence , Crystallography, X-Ray , Evolution, Molecular , Introns/genetics , Molecular Sequence Data , Nucleotide Motifs/genetics , Structure-Activity Relationship
10.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 7): 751-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22751660

ABSTRACT

Succinyl-CoA synthetase (SCS) from Thermus aquaticus was characterized biochemically via measurements of the activity of the enzyme and determination of its quaternary structure as well as its stability and refolding properties. The enzyme is most active between pH 8.0 and 8.4 and its activity increases with temperature to about 339 K. Gel-filtration chromatography and sedimentation equilibrium under native conditions demonstrated that the enzyme is a heterotetramer of two α-subunits and two ß-subunits. The activity assays showed that the enzyme uses either ADP/ATP or GDP/GTP, but prefers GDP/GTP. This contrasts with Escherichia coli SCS, which uses GDP/GTP but prefers ADP/ATP. To understand the nucleotide preference, T. aquaticus SCS was crystallized in the presence of GDP, leading to the determination of the structure in complex with GDP-Mn(2+). A water molecule and Pro20ß in T. aquaticus take the place of Gln20ß in pig GTP-specific SCS, interacting well with the guanine base and other residues of the nucleotide-binding site. This leads to the preference for GDP/GTP, but does not hinder the binding of ADP/ATP.


Subject(s)
Guanosine Triphosphate/metabolism , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Thermus/enzymology , Catalytic Domain , Crystallography, X-Ray , Enzyme Stability , Guanosine Diphosphate/metabolism , Manganese/metabolism , Models, Molecular , Protein Refolding , Protein Structure, Quaternary , Thermus/chemistry , Thermus/metabolism
11.
J Biol Chem ; 287(34): 28943-55, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22740690

ABSTRACT

Mutations in the erythroid-specific aminolevulinic acid synthase gene (ALAS2) cause X-linked sideroblastic anemia (XLSA) by reducing mitochondrial enzymatic activity. Surprisingly, a patient with the classic XLSA phenotype had a novel exon 11 mutation encoding a recombinant enzyme (p.Met567Val) with normal activity, kinetics, and stability. Similarly, both an expressed adjacent XLSA mutation, p.Ser568Gly, and a mutation (p.Phe557Ter) lacking the 31 carboxyl-terminal residues also had normal or enhanced activity, kinetics, and stability. Because ALAS2 binds to the ß subunit of succinyl-CoA synthetase (SUCLA2), the mutant proteins were tested for their ability to bind to this protein. Wild type ALAS2 bound strongly to a SUCLA2 affinity column, but the adjacent XLSA mutant enzymes and the truncated mutant did not bind. In contrast, vitamin B6-responsive XLSA mutations p.Arg452Cys and p.Arg452His, with normal in vitro enzyme activity and stability, did not interfere with binding to SUCLA2 but instead had loss of positive cooperativity for succinyl-CoA binding, an increased K(m) for succinyl-CoA, and reduced vitamin B6 affinity. Consistent with the association of SUCLA2 binding with in vivo ALAS2 activity, the p.Met567GlufsX2 mutant protein that causes X-linked protoporphyria bound strongly to SUCLA2, highlighting the probable role of an ALAS2-succinyl-CoA synthetase complex in the regulation of erythroid heme biosynthesis.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic , Genetic Diseases, X-Linked , Mutation, Missense , Succinate-CoA Ligases/metabolism , 5-Aminolevulinate Synthetase/genetics , Adult , Amino Acid Substitution , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/genetics , Enzyme Stability/genetics , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , Heme/biosynthesis , Heme/genetics , Humans , Male , Protein Binding , Protoporphyria, Erythropoietic/enzymology , Protoporphyria, Erythropoietic/genetics , Succinate-CoA Ligases/genetics , Vitamin B 6/genetics , Vitamin B 6/metabolism
12.
Arch Biochem Biophys ; 521(1-2): 51-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22446164

ABSTRACT

Substrate initially binds to ß-galactosidase (Escherichia coli) at a 'shallow' site. It then moves ∼3Å to a 'deep' site and the transition state forms. Asn460 interacts in both sites, forming a water bridge interaction with the O3 hydroxyl of the galactosyl moiety in the shallow site and a direct H-bond with the O2 hydroxyl of the transition state in the deep site. Structural and kinetic studies were done with ß-galactosidases with substitutions for Asn460. The substituted enzymes have enhanced substrate affinity in the shallow site indicating lower E·substrate complex energy levels. They have poor transition state stabilization in the deep site that is manifested by increased energy levels of the E·transition state complexes. These changes in stability result in increased activation energies and lower k(cat) values. Substrate affinity to N460D-ß-galactosidase was enhanced through greater binding enthalpy (stronger H-bonds through the bridging water) while better affinity to N460T-ß-galactosidase occurred because of greater binding entropy. The transition states are less stable with N460S- and N460T-ß-galactosidase because of the weakening or loss of the important bond to the O2 hydroxyl of the transition state. For N460D-ß-galactosidase, the transition state is less stable due to an increased entropy penalty.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Amino Acid Substitution , Catalytic Domain/genetics , Crystallography, X-Ray , Enzyme Activation , Enzyme Stability/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Hydrogen Bonding , Kinetics , Models, Molecular , Substrate Specificity/genetics , Thermodynamics , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/genetics
13.
Arch Biochem Biophys ; 517(2): 111-22, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22155115

ABSTRACT

A loop (residues 794-803) at the active site of ß-galactosidase (Escherichia coli) opens and closes during catalysis. The α and ß carbons of Ser-796 form a hydrophobic connection to Phe-601 when the loop is closed while a connection via two H-bonds with the Ser hydroxyl occurs with the loop open. ß-Galactosidases with substitutions for Ser-796 were investigated. Replacement by Ala strongly stabilizes the closed conformation because of greater hydrophobicity and loss of H-bonding ability while replacement with Thr stabilizes the open form through hydrophobic interactions with its methyl group. Upon substitution with Asp much of the defined loop structure is lost. The different open-closed equilibria cause differences in the stabilities of the enzyme·substrate and enzyme·transition state complexes and of the covalent intermediate that affect the activation thermodynamics. With Ala, large changes of both the galactosylation (k(2)) and degalactosylation (k(3)) rates occur. With Thr and Asp, the k(2) and k(3) were not changed as much but large ΔH(‡) and TΔS(‡) changes showed that the substitutions caused mechanistic changes. Overall, the hydrophobic and H-bonding properties of Ser-796 result in interactions strong enough to stabilize the open or closed conformations of the loop but weak enough to allow loop movement during the reaction.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Amino Acid Substitution , Catalytic Domain/genetics , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Isopropyl Thiogalactoside/pharmacology , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nitrophenylgalactosides/pharmacology , Protein Conformation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Static Electricity , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/genetics
14.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 10): 1168-72, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-22102020

ABSTRACT

Human ATP-citrate lyase (EC 2.3.3.8) is the cytoplasmic enzyme that catalyzes the production of acetyl-CoA from citrate, CoA and ATP. The amino-terminal portion of the enzyme, containing residues 1-817, was crystallized in the presence of tartrate, ATP and magnesium ions. The crystals diffracted to 2.3 Å resolution. The structure shows ADP-Mg(2+) bound to the domain that possesses the ATP-grasp fold. The structure demonstrates that this crystal form could be used to investigate the structures of complexes with inhibitors of ATP-citrate lyase that bind at either the citrate- or ATP-binding site.


Subject(s)
ATP Citrate (pro-S)-Lyase/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Magnesium/chemistry , Protein Interaction Domains and Motifs , ATP Citrate (pro-S)-Lyase/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Crystallography, X-Ray , Escherichia coli/enzymology , Humans , Magnesium/metabolism , Models, Molecular , Structural Homology, Protein
15.
Biochemistry ; 49(48): 10319-28, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-20977214

ABSTRACT

Catalysis by succinyl-CoA:3-oxoacid CoA transferase proceeds through a thioester intermediate in which CoA is covalently linked to the enzyme. To determine the conformation of the thioester intermediate, crystals of the pig enzyme were grown in the presence of the substrate acetoacetyl-CoA. X-ray diffraction data show the enzyme in both the free form and covalently bound to CoA via Glu305. In the complex, the protein adopts a conformation in which residues 267-275, 280-287, 357-373, and 398-477 have shifted toward Glu305, closing the enzyme around the thioester. Enzymes provide catalysis by stabilizing the transition state relative to complexes with substrates or products. In this case, the conformational change allows the enzyme to interact with parts of CoA distant from the reactive thiol while the thiol is covalently linked to the enzyme. The enzyme forms stabilizing interactions with both the nucleotide and pantoic acid portions of CoA, while the interactions with the amide groups of the pantetheine portion are poor. The results shed light on how the enzyme uses the binding energy for groups remote from the active center of CoA to destabilize atoms closer to the active center, leading to acceleration of the reaction by the enzyme.


Subject(s)
Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Biocatalysis , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Stability/drug effects , Kinetics , Models, Molecular , Protein Binding , Protein Conformation/drug effects , Swine
16.
J Biol Chem ; 285(35): 27418-27428, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20558738

ABSTRACT

ATP-citrate lyase (ACLY) catalyzes the conversion of citrate and CoA into acetyl-CoA and oxaloacetate, coupled with the hydrolysis of ATP. In humans, ACLY is the cytoplasmic enzyme linking energy metabolism from carbohydrates to the production of fatty acids. In situ proteolysis of full-length human ACLY gave crystals of a truncated form, revealing the conformations of residues 2-425, 487-750, and 767-820 of the 1101-amino acid protein. Residues 2-425 form three domains homologous to the beta-subunit of succinyl-CoA synthetase (SCS), while residues 487-820 form two domains homologous to the alpha-subunit of SCS. The crystals were grown in the presence of tartrate or the substrate, citrate, and the structure revealed the citrate-binding site. A loop formed by residues 343-348 interacts via specific hydrogen bonds with the hydroxyl and carboxyl groups on the prochiral center of citrate. Arg-379 forms a salt bridge with the pro-R carboxylate of citrate. The pro-S carboxylate is free to react, providing insight into the stereospecificity of ACLY. Because this is the first structure of any member of the acyl-CoA synthetase (NDP-forming) superfamily in complex with its organic acid substrate, locating the citrate-binding site is significant for understanding the catalytic mechanism of each member, including the prototype SCS. Comparison of the CoA-binding site of SCSs with the similar structure in ACLY showed that ACLY possesses a different CoA-binding site. Comparisons of the nucleotide-binding site of SCSs with the similar structure in ACLY indicates that this is the ATP-binding site of ACLY.


Subject(s)
ATP Citrate (pro-S)-Lyase/chemistry , Adenosine Triphosphate/chemistry , Coenzyme A/chemistry , Cytoplasm/enzymology , Tartrates/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Humans , Structure-Activity Relationship
17.
Protein Sci ; 18(5): 1128-34, 2009 May.
Article in English | MEDLINE | ID: mdl-19388079

ABSTRACT

Recent X-ray crystal structures and solution NMR spectroscopy data for calcium- and integrin-binding protein 1 (CIB1) have all revealed a common EF-hand domain structure for the protein. However, the orientation of the two protein domains, the oligomerization state, and the conformations of the N- and C-terminal extensions differ among the structures. In this study, we examine whether the binding of glutathione or auxiliary Ca(2+) ions as observed in the crystal structures, occur in solution, and whether these interactions can influence the structure or dimerization of CIB1. In addition, we test the potential phosphatase activity of CIB1, which was hypothesized based on the glutathione binding site geometry observed in one of the crystal structures of the protein. Biophysical and biochemical experiments failed to detect glutathione binding, protein dimerization, or phosphatase activity for CIB1 under several solution conditions. However, our data identify low affinity (K(d), 10(-2)M) Ca(2+) binding events that influence the structures of the N- and C-terminal extensions of CIB1 under high (300 mM) Ca(2+) crystallization conditions. In addition to providing a rationale for differences amongst the various solution and crystal structures of CIB1, our results show that the impact of low affinity Ca(2+) binding events should be considered when analyzing and interpreting protein crystallographic structures determined in the presence of very high Ca(2+) concentrations.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium/metabolism , Binding Sites , Calcium-Binding Proteins/metabolism , Crystallography, X-Ray , Glutathione/metabolism , Light , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Scattering, Radiation
18.
Biochemistry ; 46(38): 10852-63, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17718512

ABSTRACT

Succinyl-CoA:3-ketoacid CoA transferase (SCOT) transfers CoA from succinyl-CoA to acetoacetate via a thioester intermediate with its active site glutamate residue, Glu 305. When CoA is linked to the enzyme, a cysteine residue can now be rapidly modified by 5,5'-dithiobis(2-nitrobenzoic acid), reflecting a conformational change of SCOT upon formation of the thioester. Since either Cys 28 or Cys 196 could be the target, each was mutated to Ser to distinguish between them. Like wild-type SCOT, the C196S mutant protein was modified rapidly in the presence of acyl-CoA substrates. In contrast, the C28S mutant protein was modified much more slowly under identical conditions, indicating that Cys 28 is the residue exposed on binding CoA. The specific activity of the C28S mutant protein was unexpectedly lower than that of wild-type SCOT. X-ray crystallography revealed that Ser adopts a different conformation than the native Cys. A chloride ion is bound to one of four active sites in the crystal structure of the C28S mutant protein, mimicking substrate, interacting with Lys 329, Asn 51, and Asn 52. On the basis of these results and the studies of the structurally similar CoA transferase from Escherichia coli, YdiF, bound to CoA, the conformational change in SCOT was deduced to be a domain rotation of 17 degrees coupled with movement of two loops: residues 321-329 that bury Cys 28 and interact with succinate or acetoacetate and residues 374-386 that interact with CoA. Modeling this conformational change has led to the proposal of a new mechanism for catalysis by SCOT.


Subject(s)
Amino Acid Substitution , Coenzyme A-Transferases/chemistry , Coenzyme A/metabolism , Cysteine/chemistry , Myocardium/enzymology , Amino Acid Sequence , Animals , Binding Sites , Coenzyme A/chemistry , Coenzyme A-Transferases/isolation & purification , Coenzyme A-Transferases/metabolism , Crystallography, X-Ray/methods , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Glutamic Acid/chemistry , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid , Serine/chemistry , Swine
19.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 8): 876-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17642514

ABSTRACT

Succinyl-CoA synthetase has a highly conserved cysteine residue, Cys123alpha in the Escherichia coli enzyme, that is located near the CoA-binding site and the active-site histidine residue. To test whether the succinyl moiety of succinyl-CoA is transferred to the thiol of Cys123alpha as part of the catalytic mechanism, this residue was mutated to alanine, serine, threonine and valine. Each mutant protein was catalytically active, although less active than the wild type. This proved that the specific formation of a thioester bond with Cys123alpha is not part of the catalytic mechanism. To understand why the mutations affected catalysis, the crystal structures of the four mutant proteins were determined. The alanine mutant showed no structural changes yet had reduced activity, suggesting that the size of the cysteine is important for optimal activity. These results explain why this cysteine residue is conserved in the sequences of succinyl-CoA synthetases from different sources.


Subject(s)
Cysteine/metabolism , Escherichia coli/enzymology , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Catalysis , Crystallography, X-Ray , Cysteine/genetics , Escherichia coli/genetics , Histidine/analogs & derivatives , Histidine/chemistry , Histidine/metabolism , Kinetics , Models, Molecular , Mutation/genetics , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Succinate-CoA Ligases/genetics , Temperature
20.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 63(Pt 5): 399-402, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17565180

ABSTRACT

Succinyl-CoA synthetase (SCS) is an enzyme of the citric acid cycle and is thus found in most species. To date, there are no structures available of SCS from a thermophilic organism. To investigate how the enzyme adapts to higher temperatures, SCS from Thermus aquaticus was cloned, overexpressed, purified and crystallized. Attempts to crystallize the enzyme were thwarted by proteolysis of the beta-subunit and preferential crystallization of the truncated form. Crystals of full-length SCS were grown after the purification protocol was modified to include frequent additions of protease inhibitors. The resulting crystals, which diffract to 2.35 A resolution, are of the protein in complex with Mn2+-GDP.


Subject(s)
Acyl Coenzyme A/chemistry , Ligases/chemistry , Thermus/enzymology , Base Sequence , Cloning, Molecular , Crystallization , Crystallography, X-Ray , DNA Primers , Ligases/genetics , Ligases/isolation & purification , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...