Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Publication year range
1.
Cell ; 158(6): 1389-1401, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215494

ABSTRACT

Cyclic di-adenosine monophosphate (c-di-AMP) is a broadly conserved second messenger required for bacterial growth and infection. However, the molecular mechanisms of c-di-AMP signaling are still poorly understood. Using a chemical proteomics screen for c-di-AMP-interacting proteins in the pathogen Listeria monocytogenes, we identified several broadly conserved protein receptors, including the central metabolic enzyme pyruvate carboxylase (LmPC). Biochemical and crystallographic studies of the LmPC-c-di-AMP interaction revealed a previously unrecognized allosteric regulatory site 25 Å from the active site. Mutations in this site disrupted c-di-AMP binding and affected catalytic activity of LmPC as well as PC from pathogenic Enterococcus faecalis. C-di-AMP depletion resulted in altered metabolic activity in L. monocytogenes. Correction of this metabolic imbalance rescued bacterial growth, reduced bacterial lysis, and resulted in enhanced bacterial burdens during infection. These findings greatly expand the c-di-AMP signaling repertoire and reveal a central metabolic regulatory role for a cyclic dinucleotide.


Subject(s)
Dinucleoside Phosphates/metabolism , Listeria monocytogenes/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Bacteriolysis , Binding Sites , Crystallography, X-Ray , Host-Pathogen Interactions , Listeria monocytogenes/enzymology , Listeria monocytogenes/growth & development , Listeriosis/microbiology , Mice , Models, Molecular , Molecular Sequence Data
2.
Mol Genet Metab ; 139(2): 107605, 2023 06.
Article in English | MEDLINE | ID: mdl-37207470

ABSTRACT

Pyruvate carboxylase (PC) deficiency is a rare autosomal recessive mitochondrial neurometabolic disorder of energy deficit resulting in high morbidity and mortality, with limited therapeutic options. The PC homotetramer has a critical role in gluconeogenesis, anaplerosis, neurotransmitter synthesis, and lipogenesis. The main biochemical and clinical findings in PC deficiency (PCD) include lactic acidosis, ketonuria, failure to thrive, and neurological dysfunction. Use of the anaplerotic agent triheptanoin on a limited number of individuals with PCD has had mixed results. We expand on the potential utility of triheptanoin in PCD by examining the clinical, biochemical, molecular, and health-related quality-of-life (HRQoL) findings in a cohort of 12 individuals with PCD (eight with Type A and two each with Types B and C) treated with triheptanoin ranging for 6 days to about 7 years. The main endpoints were changes in blood lactate and HRQoL scores, but collection of useful data was limited to about half of subjects. An overall trend of lactate reduction with time on triheptanoin was noted, but with significant variability among subjects and only one subject reaching close to statistical significance for this endpoint. Parent reported HRQoL assessments with treatment showed mixed results, with some subjects showing no change, some improvement, and some worsening of overall scores. Subjects with buried amino acids in the pyruvate carboxyltransferase domain of PC that undergo destabilizing replacements may be more likely to respond (with lactate reduction or HRQoL improvement) to triheptanoin compared to those with replacements that disrupt tetramerization or subunit-subunit interface contacts. The reason for this difference is unclear and requires further validation. We observed significant variability but an overall trend of lactate reduction with time on triheptanoin and mixed parent reported outcome changes by HRQoL assessments for subjects with PCD on long-term triheptanoin. The mixed results noted with triheptanoin therapy in this study could be due to endpoint data limitation, variability of disease severity between subjects, limitation of the parent reported HRQoL tool, or subject genotype variability. Alternative designed trials and more study subjects with PCD will be needed to validate important observations from this work.


Subject(s)
Pyruvate Carboxylase Deficiency Disease , Humans , Pyruvate Carboxylase Deficiency Disease/drug therapy , Pyruvate Carboxylase Deficiency Disease/genetics , Triglycerides , Mitochondria , Lactates , Pyruvate Carboxylase/genetics , Pyruvate Carboxylase/chemistry
3.
Biochemistry ; 61(17): 1824-1835, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35943735

ABSTRACT

Biotin-dependent enzymes employ a carrier domain to efficiently transport reaction intermediates between distant active sites. The translocation of this carrier domain is critical to the interpretation of kinetic and structural studies, but there have been few direct attempts to investigate the dynamic interplay between ligand binding and carrier domain positioning in biotin-dependent enzymes. Pyruvate carboxylase (PC) catalyzes the MgATP-dependent carboxylation of pyruvate where the biotinylated carrier domain must translocate ∼70 Šfrom the biotin carboxylase domain to the carboxyltransferase domain. Many prior studies have assumed that carrier domain movement is governed by ligand-induced conformational changes, but the mechanism underlying this movement has not been confirmed. Here, we have developed a system to directly observe PC carrier domain positioning in both the presence and absence of ligands, independent of catalytic turnover. We have incorporated a cross-linking trap that reports on the interdomain conformation of the carrier domain when it is positioned in proximity to a neighboring carboxyltransferase domain. Cross-linking was monitored by gel electrophoresis, inactivation kinetics, and intrinsic tryptophan fluorescence. We demonstrate that the carrier domain positioning equilibrium is sensitive to substrate analogues and the allosteric activator acetyl-CoA. Notably, saturating concentrations of biotin carboxylase ligands do not prevent carrier domain trapping proximal to the neighboring carboxyltransferase domain, demonstrating that carrier domain positioning is governed by conformational selection. This model of carrier domain translocation in PC can be applied to other multi-domain enzymes that employ large-scale domain motions to transfer intermediates during catalysis.


Subject(s)
Carboxyl and Carbamoyl Transferases , Pyruvate Carboxylase , Acetyl-CoA Carboxylase/metabolism , Biotin/chemistry , Carboxyl and Carbamoyl Transferases/genetics , Carboxyl and Carbamoyl Transferases/metabolism , Catalytic Domain , Ligands , Pyruvate Carboxylase/chemistry , Staphylococcus aureus
4.
Arch Biochem Biophys ; 711: 109017, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34411580

ABSTRACT

A previous study showed that 2'-3'-O-(2,4,6-trinitrophenyl) adenosine 5'-triphosphate (TNP-ATP) was a weak allosteric activator of Rhizobium etli pyruvate carboxylase (RePC) in the absence of acetyl-CoA. On the other hand, TNP-ATP inhibited the allosteric activation of RePC by acetyl-CoA. Here, we aimed to study the role of triphosphate group of TNP-ATP on its allosteric activation of the enzyme and inhibition of acetyl-CoA-dependent activation of RePC using TNP-ATP and its derivatives, including TNP-ADP, TNP-AMP and TNP-adenosine. The pyruvate carboxylation activity was assayed to determine the effect of reducing the number of phosphate groups in TNP-ATP derivatives on allosteric activation and inhibition of acetyl-CoA activation of RePC and chicken liver pyruvate carboxylase (CLPC). Reducing the number of phosphate groups in TNP-ATP derivatives decreased the activation efficacy for both RePC and CLPC compared to TNP-ATP. The apparent binding affinity and inhibition of activation of the enzymes by acetyl-CoA were also diminished when the number of phosphate groups in the TNP-ATP derivatives was reduced. Whilst TNP-AMP activated RePC, it did not activate CLPC, but it did inhibit acetyl-CoA activation of both RePC and CLPC. Similarly, TNP-adenosine did not activate RePC; however, it did inhibit acetyl-CoA activation using a different mechanism compared to phosphorylated TNP-derivatives. These findings indicate that mechanisms of PC activation and inhibition of acetyl-CoA activation by TNP-ATP and its derivatives are different. This study provides the basis for possible drug development for treatment of metabolic diseases and cancers with aberrant expression of PC.


Subject(s)
Acetyl Coenzyme A/chemistry , Adenosine Triphosphate/analogs & derivatives , Allosteric Regulation/drug effects , Enzyme Activators/chemistry , Pyruvate Carboxylase/chemistry , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Triphosphate/chemistry , Animals , Chickens , Enzyme Assays , Kinetics , Liver/enzymology , Molecular Structure
5.
Arch Biochem Biophys ; 695: 108630, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33080172

ABSTRACT

The formation, kinetics and thermodynamic activation parameters of hybrid tetramers of pyruvate carboxylase (PC) formed between wild-type Rhizobium etli pyruvate carboxylase (WTRePC) and mutant forms of this enzyme, as well as between Aspergillus nidulans PC and mutant forms of RePC have been characterized in a previous study. In this current work, we aim to extend the previous study by forming hybrid tetramers between WTRePC or chicken liver PC (CLPC) with single or double mutant RePCs. By forming hybrid tetramers between WTRePC with either K1119A or ΔBCCP RePC, the biotin moiety and BCCP (biotin carboxyl carrier protein) domain appear to play a crucial role in determination of thermodynamic activation parameters, especially the activation entropy, and the order of tetrameric structure. Using E218A:K1119A hybrid tetramers, an alternative pathway of biotin carboxylation occurred only in the absence of acetyl CoA. In this pathway, the biotin of the E218A subunits is carboxylated in the BC domain of the K1119A subunits, since the E218A mutation destroys the catalytic activity of the BC domain. Transfer of the carboxyl group to pyruvate could then occur in the CT domain of either E218A or K1119A. Part of the reduction of activity in hybrid tetramers of WTRePC and double mutant, E218A.K1119A could result from the loss of this pathway. Previously, D1018A mutant RePC homotetramers exhibited a 12-fold increase in the rate constant for catalysis in the absence of acetyl CoA. This was taken to indicate that inter-residue interactions involving D1018 inhibit the interconversion between the symmetrical and asymmetrical forms of the tetramer in the absence of acetyl CoA. The mutation, D1018A, in hybrid tetramers of WTRePC:D1018A.K1119A (D1018A.K1119A is a double mutant form of RePC) had no such effect on the rate constant, suggesting that in hybrid tetramers obligatory oscillation between asymmetrical and symmetrical conformers of the tetramer is not required to drive the catalytic cycle. Finally, K1119A or E218A RePC mutant can form hybrid tetramers with PC subunits from an evolutionarily distant species, chicken, that have stability characteristics that lie between those of the homotetramers of the two enzymes. This work provides insights into the how the PC tetramer functions to perform catalysis and is regulated by acetyl CoA. The ability to form hybrid tetrameric PCs composed of PC subunits from widely varying species that have a mixture of characteristics of the two source enzymes may also provide ways of developing novel PCs for biotechnological purposes.


Subject(s)
Aspergillus nidulans , Avian Proteins/chemistry , Bacterial Proteins/chemistry , Biotin/chemistry , Chickens , Fungal Proteins/chemistry , Liver/enzymology , Pyruvate Carboxylase/chemistry , Rhizobium etli , Animals , Aspergillus nidulans/enzymology , Aspergillus nidulans/genetics , Avian Proteins/genetics , Avian Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biotin/genetics , Biotin/metabolism , Catalysis , Fungal Proteins/genetics , Fungal Proteins/metabolism , Protein Domains , Protein Structure, Quaternary , Pyruvate Carboxylase/genetics , Pyruvate Carboxylase/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhizobium etli/enzymology , Rhizobium etli/genetics
6.
Hum Mutat ; 40(6): 816-827, 2019 06.
Article in English | MEDLINE | ID: mdl-30870574

ABSTRACT

Pyruvate carboxylase deficiency (PCD) is caused by biallelic mutations of the PC gene. The reported clinical spectrum includes a neonatal form with early death (type B), an infantile fatal form (type A), and a late-onset form with isolated mild intellectual delay (type C). Apart from homozygous stop-codon mutations leading to type B PCD, a genotype-phenotype correlation has not otherwise been discernible. Indeed, patients harboring biallelic heterozygous variants leading to PC activity near zero can present either with a fatal infantile type A or with a benign late onset type C form. In this study, we analyzed six novel patients with type A (three) and type C (three) PCD, and compared them with previously reported cases. First, we observed that type C PCD is not associated to homozygous variants in PC. In silico modeling was used to map former and novel variants associated to type A and C PCD, and to predict their potential effects on the enzyme structure and function. We found that variants lead to type A or type C phenotype based on the destabilization between the two major enzyme conformers. In general, our study on novel and previously reported patients improves the overall understanding on type A and C PCD.


Subject(s)
Mutation , Pyruvate Carboxylase Deficiency Disease/genetics , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Child , Child, Preschool , Enzyme Stability , Female , Genetic Association Studies , Humans , Infant , Male , Models, Molecular , Protein Conformation , Pyruvate Carboxylase Deficiency Disease/classification , Structural Homology, Protein
7.
Arch Biochem Biophys ; 665: 87-95, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30831071

ABSTRACT

In sedimentation velocity experiments, we have been able to detect hybrid Rhizobium etli pyruvate carboxylase tetramers formed between subunits that contain covalently bound biotin and mutant subunits that do not. This was performed by forming complexes of the tetramers with the biotin-binding protein avidin. In addition, we have shown that it is possible to form hybrid tetramers of pyruvate carboxylase subunits from two different organisms (bacteria - Rhizobium etli and fungi - Aspergillus nidulans). In hybrid tetramers containing mutant subunits that are not fully catalytically active and fully catalytically active subunits, the catalytic and regulatory properties of these hybrid tetramers are modified compared to homotetramers of the fully active pyruvate carboxylase subunits. Our data indicates that the model of catalysis involving half-of-the-sites activity in which there is obligatory alternation of pyruvate carboxylating activity between pairs of subunits either face of the tetramer, does not occur in the hybrid tetramers. Our results are also discussed in relation to recent findings that there are multiple pathways of biotin carboxylation and decarboxylation between subunits in pyruvate carboxylase tetramers.


Subject(s)
Biopolymers/metabolism , Pyruvate Carboxylase/metabolism , Thermodynamics , Allosteric Regulation , Avidin/metabolism , Biopolymers/chemistry , Catalysis , Kinetics , Pyruvate Carboxylase/chemistry , Ultracentrifugation
8.
Subcell Biochem ; 83: 291-322, 2017.
Article in English | MEDLINE | ID: mdl-28271481

ABSTRACT

Pyruvate carboxylase is a metabolic enzyme that fuels the tricarboxylic acid cycle with one of its intermediates and also participates in the first step of gluconeogenesis. This large enzyme is multifunctional, and each subunit contains two active sites that catalyze two consecutive reactions that lead to the carboxylation of pyruvate into oxaloacetate, and a binding site for acetyl-CoA, an allosteric regulator of the enzyme. Pyruvate carboxylase oligomers arrange in tetramers and covalently attached biotins mediate the transfer of carboxyl groups between distant active sites. In this chapter, some of the recent findings on pyruvate carboxylase functioning are presented, with special focus on the structural studies of the full length enzyme. The emerging picture reveals large movements of domains that even change the overall quaternary organization of pyruvate carboxylase tetramers during catalysis.


Subject(s)
Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Biocatalysis , Biotin/metabolism , Protein Structure, Quaternary
9.
Biochemistry ; 56(27): 3492-3506, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28617592

ABSTRACT

Allosteric regulation of pyruvate carboxylase (PC) activity is pivotal to maintaining metabolic homeostasis. In contrast, dysregulated PC activity contributes to the pathogenesis of numerous diseases, rendering PC a possible target for allosteric therapeutic development. Recent research efforts have focused on demarcating the role of acetyl-CoA, one of the most potent activators of PC, in coordinating catalytic events within the multifunctional enzyme. Herein, we report a kinetic and thermodynamic analysis of acetyl-CoA activation of the Staphylococcus aureus PC (SaPC)-catalyzed carboxylation of pyruvate to identify novel means by which acetyl-CoA synchronizes catalytic events within the PC tetramer. Kinetic and linked-function analysis, or thermodynamic linkage analysis, indicates that the substrates of the biotin carboxylase and carboxyl transferase domain are energetically coupled in the presence of acetyl-CoA. In contrast, both kinetic and energetic coupling between the two domains is lost in the absence of acetyl-CoA, suggesting a functional role for acetyl-CoA in facilitating the long-range transmission of substrate-induced conformational changes within the PC tetramer. Interestingly, thermodynamic activation parameters for the SaPC-catalyzed carboxylation of pyruvate are largely independent of acetyl-CoA. Our results also reveal the possibility that global conformational changes give rise to observed species-specific thermodynamic activation parameters. Taken together, our kinetic and thermodynamic results provide a possible allosteric mechanism by which acetyl-CoA coordinates catalysis within the PC tetramer.


Subject(s)
Acetyl Coenzyme A/metabolism , Bacterial Proteins/metabolism , Models, Molecular , Pyruvate Carboxylase/metabolism , Staphylococcus aureus/enzymology , Acetyl Coenzyme A/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Algorithms , Allosteric Regulation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Biocatalysis , Energy Transfer , Enzyme Activation , Enzyme Stability , Kinetics , Magnesium/chemistry , Magnesium/metabolism , Molecular Conformation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Pyruvic Acid/chemistry , Pyruvic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermodynamics
10.
Biochemistry ; 55(24): 3447-60, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27254467

ABSTRACT

Protein structure, ligand binding, and catalytic turnover contributes to the governance of catalytic events occurring at spatially distinct domains in multifunctional enzymes. Coordination of these catalytic events partially rests on the ability of spatially discrete active sites to communicate with other allosteric and active sites on the same polypeptide chain (intramolecular) or on different polypeptide chains (intermolecular) within the holoenzyme. Often, communication results in long-range effects on substrate binding or product release. For example, pyruvate binding to the carboxyl transferase (CT) domain of pyruvate carboxylase (PC) increases the rate of product release in the biotin carboxylase (BC) domain. In order to address how CT domain ligand occupancy is "sensed" by other domains, we generated functional, mixed hybrid tetramers using the E218A (inactive BC domain) and T882S (low pyruvate binding, low activity) mutant forms of PC. The apparent Ka pyruvate for the pyruvate-stimulated release of Pi catalyzed by the T882S:E218A[1:1] hybrid tetramer was comparable to the wild-type enzyme and nearly 10-fold lower than that for the T882S homotetramer. In addition, the ratio of the rates of oxaloacetate formation to Pi release for the WT:T882S[1:1] and E218A:T882S[1:1] hybrid tetramer-catalyzed reactions was 0.5 and 0.6, respectively, while the T882S homotetramer exhibited a near 1:1 coupling of the two domains, suggesting that the mechanisms coordinating catalytic events is more complicated that we initially assumed. The results presented here are consistent with an intermolecular communication mechanism, where pyruvate binding to the CT domain is "sensed" by domains on a different polypeptide chain within the tetramer.


Subject(s)
Bacterial Proteins/chemistry , Biotin/metabolism , Carbon-Nitrogen Ligases/chemistry , Carboxyl and Carbamoyl Transferases/chemistry , Pyruvate Carboxylase/chemistry , Pyruvic Acid/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Binding Sites , Carbon-Nitrogen Ligases/metabolism , Carboxyl and Carbamoyl Transferases/metabolism , Catalytic Domain , Crystallography, X-Ray , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Pyruvate Carboxylase/genetics , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism , Sequence Homology, Amino Acid
11.
Biochemistry ; 55(30): 4220-8, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27379711

ABSTRACT

The mechanism of allosteric activation of pyruvate carboxylase by acetyl CoA is not fully understood. Here we have examined the roles of residues near the acetyl CoA binding site in the allosteric activation of Rhizobium etli pyruvate carboxylase using site-directed mutagenesis. Arg429 was found to be especially important for acetyl CoA binding as substitution with serine resulted in a 100-fold increase in the Ka of acetyl CoA activation and a large decrease in the cooperativity of this activation. Asp420 and Arg424, which do not make direct contact with bound acetyl CoA, were nonetheless found to affect acetyl CoA binding when mutated, probably through changed interactions with another acetyl CoA binding residue, Arg427. Thermodynamic activation parameters for the pyruvate carboxylation reaction were determined from modified Arrhenius plots and showed that acetyl CoA acts to decrease the activation free energy of the reaction by both increasing the activation entropy and decreasing the activation enthalpy. Most importantly, mutations of Asp420, Arg424, and Arg429 enhanced the activity of the enzyme in the absence of acetyl CoA. A main focus of this work was the detailed investigation of how this increase in activity occurred in the R424S mutant. This mutation decreased the activation enthalpy of the pyruvate carboxylation reaction by an amount consistent with removal of a single hydrogen bond. It is postulated that Arg424 forms a hydrogen bonding interaction with another residue that stabilizes the asymmetrical conformation of the R. etli pyruvate carboxylase tetramer, constraining its interconversion to the symmetrical conformer that is required for catalysis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Acetyl Coenzyme A/metabolism , Allosteric Regulation , Allosteric Site/genetics , Amino Acid Sequence , Arginine/chemistry , Aspartic Acid/chemistry , Bacterial Proteins/genetics , Enzyme Activation , Glutamic Acid/chemistry , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Pyruvate Carboxylase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhizobium etli/genetics
12.
Biochemistry ; 53(27): 4455-66, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24963911

ABSTRACT

Pyruvate carboxylase (PC) catalyzes the carboxylation of pyruvate to produce oxaloacetate. Its activity is directly related to insulin release and thus PC has recently attracted great interest as a potential target for diabetes treatment. In this article, the catalytic mechanism of the carboxyl transferase domain of PC from Staphylococcus aureus was investigated by using a combined quantum-mechanical/molecular-mechanical approach. Our calculation results indicate that the catalytic reaction starts from the decarboxylation of carboxybiotin to generate an enol-BTI intermediate, followed by two consecutive proton-transfer processes (from T908 to enol-BTI and from PYR to T908). During the catalytic reaction, the main-chain amide of T908 plays a key role in catching CO2 and preventing its diffusion from the active center. A triad of residues, R571, Q575, and K741, contributes both to substrate binding and enol-pyruvate stabilization. The oxyanion hole, consisting of the side-chain hydroxyl of S911 and the side-chain amino of Q870, plays an important role in stabilizing the hydroxyl anion of BTI. The coordination of the metal cation by pyruvate is a second sphere, rather than an inner sphere, interaction, and the metal cation stabilizes the species through the medium of residue K741. The decarboxylation of carboxybiotin corresponds to the highest free energy barrier of 21.7 kcal/mol. Our results may provide useful information for both the regulation of enzyme activity and the development of related biocatalytic applications.


Subject(s)
Bacterial Proteins/chemistry , Carboxyl and Carbamoyl Transferases/chemistry , Pyruvate Carboxylase/chemistry , Staphylococcus aureus/enzymology , Biocatalysis , Cations, Divalent , Coordination Complexes/chemistry , Manganese/chemistry , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Pyruvic Acid/chemistry , Quantum Theory , Thermodynamics , Zinc/chemistry
13.
Biochemistry ; 53(6): 1051-8, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24460480

ABSTRACT

His216 is a well-conserved residue in pyruvate carboxylases and, on the basis of structures of the enzyme, appears to have a role in the binding of MgATP, forming an interaction with the 3'-hydroxyl group of the ribose ring. Mutation of this residue to asparagine results in a 9-fold increase in the Km for MgATP in its steady-state cleavage in the absence of pyruvate and a 3-fold increase in the Km for MgADP in its steady-state phosphorylation by carbamoyl phosphate. However, from single-turnover experiments of MgATP cleavage, the Kd of the enzyme·MgATP complex is essentially the same in the wild-type enzyme and H216N. Direct stopped-flow measurements of nucleotide binding and release using the fluorescent analogue FTP support these observations. However, the first-order rate constant for MgATP cleavage in the single-turnover experiments in H216N is only 0.75% of that for the wild-type enzyme, and thus, the MgATP cleavage step is rate-limiting in the steady state for H216N but not for the wild-type enzyme. Close examination of the structure of the enzyme suggested that His216 may also interact with Glu218, which in turn interacts with Glu305 to form a proton relay system involved in the deprotonation of bicarbonate. Single-turnover MgATP cleavage experiments with mutations of these two residues resulted in kinetic parameters similar to those observed in H216N. We suggest that the primary role of His216 is to coordinate the binding of MgATP and the deprotonation of bicarbonate in the reaction to form the putative carboxyphosphate intermediate by participation in a proton relay system involving Glu218 and Glu305.


Subject(s)
Adenosine Triphosphate/metabolism , Histidine/chemistry , Pyruvate Carboxylase/metabolism , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Asparagine/chemistry , Bicarbonates/pharmacology , Binding Sites , Carbamyl Phosphate/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Rhizobium etli/enzymology , Ultracentrifugation
14.
J Biol Chem ; 288(27): 19915-25, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23698000

ABSTRACT

Biotin-dependent enzymes catalyze carboxyl transfer reactions by efficiently coordinating multiple reactions between spatially distinct active sites. Pyruvate carboxylase (PC), a multifunctional biotin-dependent enzyme, catalyzes the bicarbonate- and MgATP-dependent carboxylation of pyruvate to oxaloacetate, an important anaplerotic reaction in mammalian tissues. To complete the overall reaction, the tethered biotin prosthetic group must first gain access to the biotin carboxylase domain and become carboxylated and then translocate to the carboxyltransferase domain, where the carboxyl group is transferred from biotin to pyruvate. Here, we report structural and kinetic evidence for the formation of a substrate-induced biotin binding pocket in the carboxyltransferase domain of PC from Rhizobium etli. Structures of the carboxyltransferase domain reveal that R. etli PC occupies a symmetrical conformation in the absence of the biotin carboxylase domain and that the carboxyltransferase domain active site is conformationally rearranged upon pyruvate binding. This conformational change is stabilized by the interaction of the conserved residues Asp(590) and Tyr(628) and results in the formation of the biotin binding pocket. Site-directed mutations at these residues reduce the rate of biotin-dependent reactions but have no effect on the rate of biotin-independent oxaloacetate decarboxylation. Given the conservation with carboxyltransferase domains in oxaloacetate decarboxylase and transcarboxylase, the structure-based mechanism described for PC may be applicable to the larger family of biotin-dependent enzymes.


Subject(s)
Bacterial Proteins/chemistry , Biotin/chemistry , Carboxyl and Carbamoyl Transferases/chemistry , Pyruvate Carboxylase/chemistry , Pyruvic Acid/chemistry , Rhizobium etli/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biotin/genetics , Biotin/metabolism , Carboxyl and Carbamoyl Transferases/genetics , Carboxyl and Carbamoyl Transferases/metabolism , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Pyruvate Carboxylase/genetics , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism , Rhizobium etli/chemistry
15.
Arch Biochem Biophys ; 544: 75-86, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24184447

ABSTRACT

Biotin-dependent enzymes catalyze carboxylation, decarboxylation and transcarboxylation reactions that participate in the primary metabolism of a wide range of organisms. In all cases, the overall reaction proceeds via two half reactions that take place in physically distinct active sites. In the first half-reaction, a carboxyl group is transferred to the 1-N' of a covalently tethered biotin cofactor. The tethered carboxybiotin intermediate subsequently translocates to a second active site where the carboxyl group is either transferred to an acceptor substrate or, in some bacteria and archaea, is decarboxylated to biotin and CO2 in order to power the export of sodium ions from the cytoplasm. A homologous carboxyltransferase domain is found in three enzymes that catalyze diverse overall reactions: carbon fixation by pyruvate carboxylase, decarboxylation and sodium transport by the biotin-dependent oxaloacetate decarboxylase complex, and transcarboxylation by transcarboxylase from Propionibacterium shermanii. Over the past several years, structural data have emerged which have greatly advanced the mechanistic description of these enzymes. This review assembles a uniform description of the carboxyltransferase domain structure and catalytic mechanism from recent studies of pyruvate carboxylase, oxaloacetate decarboxylase and transcarboxylase, three enzymes that utilize an analogous carboxyltransferase domain to catalyze the biotin-dependent decarboxylation of oxaloacetate.


Subject(s)
Biotin/metabolism , Carboxy-Lyases/metabolism , Animals , Carboxy-Lyases/chemistry , Carboxyl and Carbamoyl Transferases/chemistry , Carboxyl and Carbamoyl Transferases/metabolism , Humans , Models, Molecular , Protein Conformation , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism
16.
Arch Biochem Biophys ; 562: 70-9, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25157442

ABSTRACT

Pyruvate carboxylase (PC) is a biotin-dependent enzyme that catalyzes the MgATP-dependent carboxylation of pyruvate to oxaloacetate, an important anaplerotic reaction in central metabolism. During catalysis, carboxybiotin is translocated to the carboxyltransferase domain where the carboxyl group is transferred to the acceptor substrate, pyruvate. Many studies on the carboxyltransferase domain of PC have demonstrated an enhanced oxaloacetate decarboxylation activity in the presence of oxamate and it has been shown that oxamate accepts a carboxyl group from carboxybiotin during oxaloacetate decarboxylation. The X-ray crystal structure of the carboxyltransferase domain from Rhizobium etli PC reveals that oxamate is positioned in the active site in an identical manner to the substrate, pyruvate, and kinetic data are consistent with the oxamate-stimulated decarboxylation of oxaloacetate proceeding through a simple ping-pong bi bi mechanism in the absence of the biotin carboxylase domain. Additionally, analysis of truncated PC enzymes indicates that the BCCP domain devoid of biotin does not contribute directly to the enzymatic reaction and conclusively demonstrates a biotin-independent oxaloacetate decarboxylation activity in PC. These findings advance the description of catalysis in PC and can be extended to the study of related biotin-dependent enzymes.


Subject(s)
Biotin/chemistry , Carboxyl and Carbamoyl Transferases/chemistry , Oxamic Acid/chemistry , Pyruvate Carboxylase/chemistry , Rhizobium etli/enzymology , Binding Sites , Biotin/analogs & derivatives , Carbon-Nitrogen Ligases/chemistry , Catalysis , Cloning, Molecular , Crystallography, X-Ray , Lysine/analogs & derivatives , Lysine/chemistry , Models, Chemical , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Pyruvic Acid/chemistry , Rhizobium/metabolism
17.
Cell Mol Life Sci ; 70(5): 863-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22869039

ABSTRACT

Biotin-dependent carboxylases include acetyl-CoA carboxylase (ACC), propionyl-CoA carboxylase (PCC), 3-methylcrotonyl-CoA carboxylase (MCC), geranyl-CoA carboxylase, pyruvate carboxylase (PC), and urea carboxylase (UC). They contain biotin carboxylase (BC), carboxyltransferase (CT), and biotin-carboxyl carrier protein components. These enzymes are widely distributed in nature and have important functions in fatty acid metabolism, amino acid metabolism, carbohydrate metabolism, polyketide biosynthesis, urea utilization, and other cellular processes. ACCs are also attractive targets for drug discovery against type 2 diabetes, obesity, cancer, microbial infections, and other diseases, and the plastid ACC of grasses is the target of action of three classes of commercial herbicides. Deficiencies in the activities of PCC, MCC, or PC are linked to serious diseases in humans. Our understanding of these enzymes has been greatly enhanced over the past few years by the crystal structures of the holoenzymes of PCC, MCC, PC, and UC. The structures reveal unanticipated features in the architectures of the holoenzymes, including the presence of previously unrecognized domains, and provide a molecular basis for understanding their catalytic mechanism as well as the large collection of disease-causing mutations in PCC, MCC, and PC. This review will summarize the recent advances in our knowledge on the structure and function of these important metabolic enzymes.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Biotin/metabolism , Carbon-Carbon Ligases/metabolism , Carbon-Nitrogen Ligases/metabolism , Methylmalonyl-CoA Decarboxylase/metabolism , Pyruvate Carboxylase/metabolism , Acetyl-CoA Carboxylase/chemistry , Animals , Carbon-Carbon Ligases/chemistry , Carbon-Nitrogen Ligases/chemistry , Fatty Acid Synthase, Type II/chemistry , Fatty Acid Synthase, Type II/metabolism , Humans , Methylmalonyl-CoA Decarboxylase/chemistry , Models, Molecular , Protein Conformation , Pyruvate Carboxylase/chemistry
18.
Biochemistry ; 52(17): 2888-94, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23560609

ABSTRACT

Oxamate, an isosteric and isoelectronic inhibitory analogue of pyruvate, enhances the rate of enzymatic decarboxylation of oxaloacetate in the carboxyl transferase domain of pyruvate carboxylase (PC). It is unclear, though, how oxamate exerts a stimulatory effect on the enzymatic reaction. Herein, we report direct (13)C nuclear magnetic resonance (NMR) evidence that oxamate acts as a carboxyl acceptor, forming a carbamylated oxamate product and thereby accelerating the enzymatic decarboxylation reaction. (13)C NMR was used to monitor the PC-catalyzed formation of [4-(13)C]oxaloacetate and subsequent transfer of (13)CO(2) from oxaloacetate to oxamate. In the presence of oxamate, the apparent K(m) for oxaloacetate is artificially suppressed (from 15 to 4-5 µM). Interestingly, the steady-state kinetic analysis of the initial rates determined at varying concentrations of oxaloacetate and fixed concentrations of oxamate revealed initial velocity patterns inconsistent with a simple ping-pong-type mechanism. Rather, the patterns suggest the existence of an alternate decarboxylation pathway in which an unstable intermediate is formed. The steady-state kinetic analysis coupled with the normal (13)(V/K) kinetic isotope effect observed on C-4 of oxaloacetate [(13)(V/K) = 1.0117 ± 0.0005] indicates that the transfer of CO(2) from carboxybiotin to oxamate is the partially rate-limiting step of the enzymatic reaction. The catalytic mechanism proposed for the carboxylation of oxamate is similar to that proposed for the carboxylation of pyruvate, which occurs via the formation of an enol intermediate.


Subject(s)
Oxamic Acid/metabolism , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Carboxylic Acids/metabolism , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Pyruvate Carboxylase/chemistry , Substrate Specificity
19.
Biochemistry ; 52(3): 488-96, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23286247

ABSTRACT

Biotin carboxylase (BC) is a conserved component among biotin-dependent carboxylases and catalyzes the MgATP-dependent carboxylation of biotin, using bicarbonate as the CO2 donor. Studies with Escherichia coli BC have suggested long-range communication between the two active sites of a dimer, although its mechanism is not well understood. In addition, mutations in the dimer interface can produce stable monomers that are still catalytically active. A homologous dimer for the BC domain is observed in the structure of the tetrameric pyruvate carboxylase (PC) holoenzyme. We have introduced site-specific mutations into the BC domain dimer interface of Staphylococcus aureus PC (SaPC), equivalent to those used for E. coli BC, and also made chimeras replacing the SaPC BC domain with the E. coli BC subunit (EcBC chimera) or the yeast ACC BC domain (ScBC chimera). We assessed the catalytic activities of these mutants and characterized their oligomerization states by gel filtration and analytical ultracentrifugation experiments. The K442E mutant and the ScBC chimera disrupted the BC dimer and were catalytically inactive, while the F403A mutant and the EcBC chimera were still tetrameric and retained catalytic activity. The R54E mutant was also tetrameric but was catalytically inactive. Crystal structures of the R54E, F403A, and K442E mutants showed that they were tetrameric in the crystal, with conformational changes near the mutation site as well as in the tetramer organization. We have also produced the isolated BC domain of SaPC. In contrast to E. coli BC, the SaPC BC domain is monomeric in solution and catalytically inactive.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Staphylococcus aureus/enzymology , Amino Acid Substitution , Bacterial Proteins/genetics , Biocatalysis , Carbon-Nitrogen Ligases/genetics , Catalytic Domain , Chromatography, Gel , Crystallography, X-Ray , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Pyruvate Carboxylase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ultracentrifugation
20.
Biochem Biophys Res Commun ; 441(2): 377-82, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24157795

ABSTRACT

Pyruvate carboxylase (PC) is a biotin-dependent enzyme that catalyzes the MgATP- and bicarbonate-dependent carboxylation of pyruvate to oxaloacetate, an important anaplerotic reaction in central metabolism. The carboxyltransferase (CT) domain of PC catalyzes the transfer of a carboxyl group from carboxybiotin to the accepting substrate, pyruvate. It has been hypothesized that the reactive enolpyruvate intermediate is stabilized through a bidentate interaction with the metal ion in the CT domain active site. Whereas bidentate ligands are commonly observed in enzymes catalyzing reactions proceeding through an enolpyruvate intermediate, no bidentate interaction has yet been observed in the CT domain of PC. Here, we report three X-ray crystal structures of the Rhizobium etli PC CT domain with the bound inhibitors oxalate, 3-hydroxypyruvate, and 3-bromopyruvate. Oxalate, a stereoelectronic mimic of the enolpyruvate intermediate, does not interact directly with the metal ion. Instead, oxalate is buried in a pocket formed by several positively charged amino acid residues and the metal ion. Furthermore, both 3-hydroxypyruvate and 3-bromopyruvate, analogs of the reaction product oxaloacetate, bind in an identical manner to oxalate suggesting that the substrate maintains its orientation in the active site throughout catalysis. Together, these structures indicate that the substrates, products and intermediates in the PC-catalyzed reaction are not oriented in the active site as previously assumed. The absence of a bidentate interaction with the active site metal appears to be a unique mechanistic feature among the small group of biotin-dependent enzymes that act on α-keto acid substrates.


Subject(s)
Carboxyl and Carbamoyl Transferases/chemistry , Plant Proteins/chemistry , Pyruvate Carboxylase/chemistry , Rhizobium etli/enzymology , Carboxyl and Carbamoyl Transferases/antagonists & inhibitors , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Oxalates/chemistry , Protein Structure, Tertiary , Pyruvate Carboxylase/antagonists & inhibitors , Pyruvates/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL