Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 589(16): 2073-9, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26149215

ABSTRACT

We have examined the roles of Asp1018, Glu1027, Arg469 and Asp471 in the allosteric domain of Rhizobium etli pyruvate carboxylase. Arg469 and Asp471 interact directly with the allosteric activator acetyl coenzyme A (acetyl CoA) and the R469S and R469K mutants showed increased enzymic activity in the presence and absence of acetyl CoA, whilst the D471A mutant exhibited no acetyl CoA-activation. E1027A, E1027R and D1018A mutants had increased activity in the absence of acetyl CoA, but not in its presence. These results suggest that most of these residues impose restrictions on the structure and/or dynamics of the enzyme to affect activity.


Subject(s)
Acetyl Coenzyme A/metabolism , Bacterial Proteins/metabolism , Models, Molecular , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Acetyl Coenzyme A/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Substitution , Arginine/chemistry , Aspartic Acid/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bicarbonates/chemistry , Biocatalysis , Glutamic Acid/chemistry , Kinetics , Magnesium/chemistry , Molecular Conformation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Stability , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Pyruvic Acid/chemistry , Pyruvic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rhizobium etli/metabolism
2.
PLoS One ; 10(6): e0129848, 2015.
Article in English | MEDLINE | ID: mdl-26070193

ABSTRACT

Pyruvate carboxylase (PC) is an anaplerotic enzyme that catalyzes the carboxylation of pyruvate to oxaloacetate, which is crucial for replenishing tricarboxylic acid cycle intermediates when they are used for biosynthetic purposes. We examined the expression of PC by immunohistochemistry of paraffin-embedded breast tissue sections of 57 breast cancer patients with different stages of cancer progression. PC was expressed in the cancerous areas of breast tissue at higher levels than in the non-cancerous areas. We also found statistical association between the levels of PC expression and tumor size and tumor stage (P < 0.05). The involvement of PC with these two parameters was further studied in four breast cancer cell lines with different metastatic potentials; i.e., MCF-7, SKBR3 (low metastasis), MDA-MB-435 (moderate metastasis) and MDA-MB-231 (high metastasis). The abundance of both PC mRNA and protein in MDA-MB-231 and MDA-MB-435 cells was 2-3-fold higher than that in MCF-7 and SKBR3 cells. siRNA-mediated knockdown of PC expression in MDA-MB-231 and MDA-MB-435 cells resulted in a 50% reduction of cell proliferation, migration and in vitro invasion ability, under both glutamine-dependent and glutamine-depleted conditions. Overexpression of PC in MCF-7 cells resulted in a 2-fold increase in their proliferation rate, migration and invasion abilities. Taken together the above results suggest that anaplerosis via PC is important for breast cancer cells to support their growth and motility.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Pyruvate Carboxylase/metabolism , Up-Regulation , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness , Pyruvate Carboxylase/genetics
3.
Biochemistry ; 53(45): 7100-6, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25330457

ABSTRACT

L-aspartate is a regulatory feedback inhibitor of the biotin-dependent enzyme pyruvate carboxylase in response to increased levels of tricarboxylic acid cycle intermediates. Detailed studies of L-aspartate inhibition of pyruvate carboxylase have been mainly confined to eukaryotic microbial enzymes, and aspects of its mode of action remain unclear. Here we examine its inhibition of the bacterial enzyme Rhizobium etli pyruvate carboxylase. Kinetic studies demonstrated that L-aspartate binds to the enzyme cooperatively and inhibits the enzyme competitively with respect to acetyl-CoA. L-aspartate also inhibits activation of the enzyme by MgTNP-ATP. The action of L-aspartate was not confined to inhibition of acetyl-CoA binding, because the acetyl-CoA-independent activity of the enzyme was also inhibited by increasing concentrations of L-aspartate. This inhibition of acetyl-CoA-independent activity was demonstrated to be focused in the biotin carboxylation domain of the enzyme, and it had no effect on the oxamate-induced oxaloacetate decarboxylation reaction that occurs in the carboxyl transferase domain. L-aspartate was shown to competitively inhibit bicarbonate-dependent MgATP cleavage with respect to MgATP but also probably inhibits carboxybiotin formation and/or translocation of the carboxybiotin to the site of pyruvate carboxylation. Unlike acetyl-CoA, L-aspartate has no effect on the coupling between MgATP cleavage and oxaloacetate formation. The results suggest that the three allosteric effector sites (acetyl-CoA, MgTNP-ATP, and L-aspartate) are spatially distinct but connected by a network of allosteric interactions.


Subject(s)
Aspartic Acid/pharmacology , Pyruvate Carboxylase/antagonists & inhibitors , Rhizobium etli/enzymology , Aspartic Acid/metabolism , Enzyme Inhibitors/pharmacology , Pyruvate Carboxylase/metabolism , Rhizobium etli/drug effects
4.
Bioorg Med Chem Lett ; 24(19): 4689-4693, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25193234

ABSTRACT

Inhibitors of Staphylococcus aureus biotin protein ligase (SaBPL) are generated by replacing the acyl phosphate group of biotinyl-5'-AMP with either a 1,2,3-triazole (see 5/10a/10b) or a 1,2,4-oxadiazole (see 7) bioisostere. Importantly, the inhibitors are inactive against the human BPL. The nature of the 5-substituent in the component benzoxazolone of the optimum 1,2,3-triazole series is critical to activity, where this group binds in the ATP binding pocket of the enzyme.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Biotin/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds/pharmacology , Ligases/antagonists & inhibitors , Organophosphates/pharmacology , Bacterial Proteins/metabolism , Biotin/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Humans , Ligases/metabolism , Models, Molecular , Molecular Structure , Organophosphates/chemical synthesis , Organophosphates/chemistry , Staphylococcus aureus/enzymology
5.
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
6.
Mol Microbiol ; 91(1): 110-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24261685

ABSTRACT

Protein biotinylation is catalysed by biotin protein ligase (BPL). The most characterized BPL is from Escherichia coli where it functions as both a biotin ligase and a homodimeric transcriptional repressor. Here we investigated another bifunctional BPL from the clinically important Staphylococcus aureus (SaBPL). Unliganded SaBPL (apo) exists in a dimer-monomer equilibrium at low micromolar concentrations - a stark contrast to E. coli BPL (EcBPL) that is monomeric under the same conditions. EMSA and SAXS analysis demonstrated that dimeric apo SaBPL adopted a conformation that was competent to bind DNA and necessary for it to function as a transcription factor. The SaBPL dimer-monomer dissociation constant was 5.8-fold tighter when binding the inhibitor biotin acetylene, but unchanged with biotin. F123, located in the dimer interface, was critical for homodimerization. Inhibition studies together with surface plasmon resonance analyses revealed a strong correlation between inhibitor potency and slow dissociation kinetics. A 24-fold difference in Ki values for these two enzymes was explained by differences in enzyme:inhibitor dissociation rates. Substitution of F123 in SaBPL and its equivalent in EcBPL altered both inhibitor potency and dissociation. Hence, F123 in SaBPL has novel roles in both protein dimerization and ligand-binding that have not been reported in EcBPL.


Subject(s)
Binding Sites/physiology , Biotin/metabolism , Ligases/chemistry , Ligases/metabolism , Phenylalanine/metabolism , Staphylococcus aureus/enzymology , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites/genetics , Biotin/antagonists & inhibitors , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ligands , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Structure, Quaternary , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Scattering, Small Angle , Staphylococcus aureus/genetics , Surface Plasmon Resonance , X-Ray Diffraction
7.
Protein Sci ; 22(6): 762-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23559560

ABSTRACT

The essential metabolic enzyme biotin protein ligase (BPL) is a potential target for the development of new antibiotics required to combat drug-resistant pathogens. Staphylococcus aureus BPL (SaBPL) is a bifunctional protein, possessing both biotin ligase and transcription repressor activities. This positions BPL as a key regulator of several important metabolic pathways. Here, we report the structural analysis of both holo- and apo-forms of SaBPL using X-ray crystallography. We also present small-angle X-ray scattering data of SaBPL in complex with its biotin-carboxyl carrier protein substrate as well as the SaBPL:DNA complex that underlies repression. This has revealed the molecular basis of ligand (biotinyl-5'-AMP) binding and conformational changes associated with catalysis and repressor function. These data provide new information to better understand the bifunctional activities of SaBPL and to inform future strategies for antibiotic discovery.


Subject(s)
Biotin/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/metabolism , Staphylococcus aureus/enzymology , Acetyl-CoA Carboxylase/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/metabolism , Amino Acid Sequence , Biotin/analogs & derivatives , Crystallography, X-Ray , Fatty Acid Synthase, Type II/metabolism , Humans , Molecular Sequence Data , Protein Interaction Maps , Protein Multimerization , Sequence Alignment , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism
8.
Hybridoma (Larchmt) ; 31(5): 305-13, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23098296

ABSTRACT

Pyruvate carboxylase [EC 6.4.1.1] plays an important anaplerotic role in many species by catalyzing the carboxylation of pyruvate to oxaloacetate. To extend our understanding about the structure and function of pyruvate carboxylase (PC), a series of monoclonal antibodies were raised against sheep liver PC and those displaying inhibitory activity were further characterized. The binding epitopes of two monoclonal antibodies that displayed strong inhibitory activity were mapped. Six overlapping fragments of the human enzyme were expressed as thioredoxin fusion proteins in Escherichia coli and subjected to Western blot analysis. Both monoclonal antibodies (MAbs) recognized fragments encompassing the enzyme's C-terminal region, known to contain the structured biotin domain. Through deletion analysis, this domain was determined to be a minimal size of 80 amino acids. Further deletions that disrupted the conformation of the domain abolished antibody binding, indicating these antibodies recognized discontinuous epitopes. To further define the critical residues required for antibody recognition, a model of the domain was produced and an alanine scan performed on selected surface-exposed residues. Our results show that residues encompassing the biotin attachment site, but not biotin itself, are critical for the binding of both antibodies. These data provide a mechanism to explain the inhibitory activity of the antibodies.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibody Specificity , Biotin/chemistry , Pyruvate Carboxylase/immunology , Alanine/chemistry , Alanine/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Binding Sites, Antibody , Binding, Competitive , Biotin/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes , Escherichia coli/genetics , Humans , Liver/enzymology , Models, Molecular , Protein Structure, Tertiary , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sheep , Thioredoxins/genetics
9.
Biochemistry ; 51(41): 8208-17, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-22985389

ABSTRACT

Mutation of Arg427 and Arg472 in Rhizobium etli pyruvate carboxylase to serine or lysine greatly increased the activation constant (K(a)) of acetyl CoA, with the increase being greater for the Arg472 mutants. These results indicate that while both these residues are involved in the binding of acetyl CoA to the enzyme, Arg472 is more important than Arg427. The mutations had substantially smaller effects on the k(cat) for pyruvate carboxylation. Part of the effects of the mutations was to increase the K(m) for MgATP and the K(a) for activation by free Mg(2+) determined at saturating acetyl CoA concentrations. The inhibitory effects of the mutations on the rates of the enzyme-catalyzed bicarbonate-dependent ATP cleavage, carboxylation of biotin, and phosphorylation of ADP by carbamoyl phosphate indicate that the major locus of the effects of the mutations was in the biotin carboxylase (BC) domain active site. Even though both Arg427 and Arg472 are distant from the BC domain active site, it is proposed that their contacts with other residues in the allosteric domain, either directly or through acetyl CoA, affect the positioning and orientation of the biotin-carboxyl carrier protein (BCCP) domain and thus the binding of biotin at the BC domain active site. On the basis of the kinetic analysis proposed here, it is proposed that mutations of Arg427 and Arg472 perturb these contacts and consequently the binding of biotin at the BC domain active site. Inhibition of pyruvate carboxylation by the allosteric inhibitor l-aspartate was largely unaffected by the mutation of either Arg427 or Arg472.


Subject(s)
Acetyl Coenzyme A/metabolism , Arginine/metabolism , Pyruvate Carboxylase/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Arginine/chemistry , Biotin/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics
10.
J Biol Chem ; 287(21): 17823-17832, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22437830

ABSTRACT

There is a well documented need to replenish the antibiotic pipeline with new agents to combat the rise of drug resistant bacteria. One strategy to combat resistance is to discover new chemical classes immune to current resistance mechanisms that inhibit essential metabolic enzymes. Many of the obvious drug targets that have no homologous isozyme in the human host have now been investigated. Bacterial drug targets that have a closely related human homologue represent a new frontier in antibiotic discovery. However, to avoid potential toxicity to the host, these inhibitors must have very high selectivity for the bacterial enzyme over the human homolog. We have demonstrated that the essential enzyme biotin protein ligase (BPL) from the clinically important pathogen Staphylococcus aureus could be selectively inhibited. Linking biotin to adenosine via a 1,2,3 triazole yielded the first BPL inhibitor selective for S. aureus BPL over the human equivalent. The synthesis of new biotin 1,2,3-triazole analogues using click chemistry yielded our most potent structure (K(i) 90 nM) with a >1100-fold selectivity for the S. aureus BPL over the human homologue. X-ray crystallography confirmed the mechanism of inhibitor binding. Importantly, the inhibitor showed cytotoxicity against S. aureus but not cultured mammalian cells. The biotin 1,2,3-triazole provides a novel pharmacophore for future medicinal chemistry programs to develop this new antibiotic class.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Biotin , Drug Resistance, Bacterial/drug effects , Enzyme Inhibitors , Ligases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Triazoles , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biotin/chemistry , Biotin/pharmacology , Cell Line , Click Chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Ligases/chemistry , Ligases/metabolism , Protein Binding , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacokinetics
11.
Biochimie ; 94(3): 608-16, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21951978

ABSTRACT

The insulin-like growth factor binding proteins are a family of six proteins (IGFBP-1 to -6) that bind insulin-like growth factors-I and -II (IGF-I/II) with high affinity. In addition to regulating IGF actions, IGFBPs have IGF-independent functions. IGFBP-2, the largest member of this family, is over-expressed in many cancers and has been proposed as a possible target for the development of novel anti-cancer therapeutics. The IGFBPs have a common architecture consisting of conserved N- and C-terminal domains joined by a variable linker domain. The solution structure and dynamics of the C-terminal domain of human IGFBP-2 have been reported (Kuang Z. et al. J. Mol. Biol. 364, 690-704, 2006) but neither the N-domain (N-BP-2) nor the linker domain have been characterised. Here we present NMR resonance assignments for human N-BP-2, achieved by recording spectra at low protein concentration using non-uniform sampling and maximum entropy reconstruction. Analysis of secondary chemical shifts shows that N-BP-2 possesses a secondary structure similar to that of other IGFBPs. Although aggregation hampered determination of the solution structure for N-BP-2, a homology model was generated based on the high degree of sequence and structure homology exhibited by the IGFBPs. This model was consistent with experimental NMR and SAXS data and displayed some unique features such as a Pro/Ala-rich non-polar insert, which formed a flexible solvent-exposed loop on the surface of the protein opposite to the IGF-binding interface. NMR data indicated that this loop could adopt either of two alternate conformations in solution - an entirely flexible conformation and one containing nascent helical structure. This loop and an adjacent poly-proline sequence may comprise a potential SH3 domain interaction site for binding to other proteins.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2/chemistry , Chromatography, Gel , Humans , Magnetic Resonance Spectroscopy , Protein Structure, Tertiary , Scattering, Small Angle
12.
J Mol Med (Berl) ; 90(1): 81-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21894551

ABSTRACT

Biotin (vitamins H and B7) is an important micronutrient as defects in its availability, metabolism or adsorption can cause serious illnesses, especially in the young. A key molecule in the biotin cycle is holocarboxylase synthetase (HLCS), which attaches biotin onto the biotin-dependent enzymes. Patients with congenital HLCS deficiency are prescribed oral biotin supplements that, in most cases, reverse the clinical symptoms. However, some patients respond poorly to biotin therapy and have an extremely poor long-term prognosis. Whilst a small number of mutations in the HLCS gene have been implicated, the molecular mechanisms that lead to the biotin-unresponsive phenotype are not understood. To improve our understanding of HLCS, limited proteolysis was performed together with yeast two-hybrid analysis. A structured domain within the N-terminal region that contained two missense mutations was identified in patients who were refractory to biotin therapy, namely p.L216R and p.L237P. Genetic studies demonstrated that the interaction between the enzyme and the protein substrate was disrupted by mutation. Further dissection of the binding mechanism using surface plasmon resonance demonstrated that the mutations reduced affinity for the substrate through a >15-fold increase in dissociation rate. Together, these data provide the first molecular explanation for HLCS-deficient patients that do not respond to biotin therapy.


Subject(s)
Biotin/metabolism , Holocarboxylase Synthetase Deficiency/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/isolation & purification , Carbon-Nitrogen Ligases/metabolism , Holocarboxylase Synthetase Deficiency/genetics , Humans , Protein Binding , Protein Interaction Mapping , Pyruvate Carboxylase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
13.
ACS Med Chem Lett ; 3(6): 509-14, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-24900501

ABSTRACT

There is a desperate need to develop new antibiotic agents to combat the rise of drug-resistant bacteria, such as clinically important Staphylococcus aureus. The essential multifunctional enzyme, biotin protein ligase (BPL), is one potential drug target for new antibiotics. We report the synthesis and characterization of a series of biotin analogues with activity against BPLs from S. aureus, Escherichia coli, and Homo sapiens. Two potent inhibitors with K i < 100 nM were identified with antibacterial activity against a panel of clinical isolates of S. aureus (MIC 2-16 µg/mL). Compounds with high ligand efficiency and >20-fold selectivity between the isozymes were identified and characterized. The antibacterial mode of action was shown to be via inhibition of BPL. The bimolecular interactions between the BPL and the inhibitors were defined by surface plasmon resonance studies and X-ray crystallography. These findings pave the way for second-generation inhibitors and antibiotics with greater potency and selectivity.

14.
PLoS One ; 6(11): e27488, 2011.
Article in English | MEDLINE | ID: mdl-22140443

ABSTRACT

BACKGROUND: Insulin-like growth factor-II (IGF-II) promotes cell proliferation and survival and plays an important role in normal fetal development and placental function. IGF-II binds both the insulin-like growth factor receptor (IGF-1R) and insulin receptor isoform A (IR-A) with high affinity. Interestingly both IGF-II and the IR-A are often upregulated in cancer and IGF-II acts via both receptors to promote cancer proliferation. There is relatively little known about the mechanism of ligand induced activation of the insulin (IR) and IGF-1R. The recently solved IR structure reveals a folded over dimer with two potential ligand binding pockets arising from residues on each receptor half. Site-directed mutagenesis has mapped receptor residues important for ligand binding to two separate sites within the ligand binding pocket and we have recently shown that the IGFs have two separate binding surfaces which interact with the receptor sites 1 and 2. METHODOLOGY/PRINCIPAL FINDINGS: In this study we describe a series of partial IGF-1R and IR agonists generated by mutating Glu12 of IGF-II. By comparing receptor binding affinities, abilities to induce negative cooperativity and potencies in receptor activation, we provide evidence that residue Glu12 bridges the two receptor halves leading to receptor activation. CONCLUSIONS/SIGNIFICANCE: This study provides novel insight into the mechanism of receptor binding and activation by IGF-II, which may be important for the future development of inhibitors of its action for the treatment of cancer.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Enzyme Activation , Enzyme Assays , Insulin-Like Growth Factor I/analogs & derivatives , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor II/analogs & derivatives , Insulin-Like Growth Factor II/chemistry , Mice , Molecular Sequence Data , Mutant Proteins/biosynthesis , Mutant Proteins/chemistry , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/chemistry , Receptor, Insulin/chemistry , Sequence Alignment , Signal Transduction
15.
Biochemistry ; 50(45): 9724-37, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21957995

ABSTRACT

The catalytic mechanism of the MgATP-dependent carboxylation of biotin in the biotin carboxylase domain of pyruvate carboxylase from R. etli (RePC) is common to the biotin-dependent carboxylases. The current site-directed mutagenesis study has clarified the catalytic functions of several residues proposed to be pivotal in MgATP-binding and cleavage (Glu218 and Lys245), HCO(3)(-) deprotonation (Glu305 and Arg301), and biotin enolization (Arg353). The E218A mutant was inactive for any reaction involving the BC domain and the E218Q mutant exhibited a 75-fold decrease in k(cat) for both pyruvate carboxylation and the full reverse reaction. The E305A mutant also showed a 75- and 80-fold decrease in k(cat) for both pyruvate carboxylation and the full reverse reaction, respectively. While Glu305 appears to be the active site base which deprotonates HCO(3)(-), Lys245, Glu218, and Arg301 are proposed to contribute to catalysis through substrate binding interactions. The reactions of the biotin carboxylase and carboxyl transferase domains were uncoupled in the R353M-catalyzed reactions, indicating that Arg353 may not only facilitate the formation of the biotin enolate but also assist in coordinating catalysis between the two spatially distinct active sites. The 2.5- and 4-fold increase in k(cat) for the full reverse reaction with the R353K and R353M mutants, respectively, suggests that mutation of Arg353 allows carboxybiotin increased access to the biotin carboxylase domain active site. The proposed chemical mechanism is initiated by the deprotonation of HCO(3)(-) by Glu305 and concurrent nucleophilic attack on the γ-phosphate of MgATP. The trianionic carboxyphosphate intermediate formed reversibly decomposes in the active site to CO(2) and PO(4)(3-). PO(4)(3-) then acts as the base to deprotonate the tethered biotin at the N(1)-position. Stabilized by interactions between the ureido oxygen and Arg353, the biotin-enolate reacts with CO(2) to give carboxybiotin. The formation of a distinct salt bridge between Arg353 and Glu248 is proposed to aid in partially precluding carboxybiotin from reentering the biotin carboxylase active site, thus preventing its premature decarboxylation prior to the binding of a carboxyl acceptor in the carboxyl transferase domain.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Adenosine Triphosphate/metabolism , Bacterial Proteins/genetics , Biotin/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Catalytic Domain , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Oxaloacetic Acid/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Pyruvate Carboxylase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhizobium etli/genetics
16.
Biochemistry ; 50(45): 9708-23, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21958016

ABSTRACT

Pyruvate carboxylase (PC) catalyzes the ATP-dependent carboxylation of pyruvate to oxaloacetate, an important anaplerotic reaction in mammalian tissues. To effect catalysis, the tethered biotin of PC must gain access to active sites in both the biotin carboxylase domain and the carboxyl transferase domain. Previous studies have demonstrated that a mutation of threonine 882 to alanine in PC from Rhizobium etli renders the carboxyl transferase domain inactive and favors the positioning of biotin in the biotin carboxylase domain. We report the 2.4 Å resolution X-ray crystal structure of the Rhizobium etli PC T882A mutant which reveals the first high-resolution description of the domain interaction between the biotin carboxyl carrier protein domain and the biotin carboxylase domain. The overall quaternary arrangement of Rhizobium etli PC remains highly asymmetrical and is independent of the presence of allosteric activator. While biotin is observed in the biotin carboxylase domain, its access to the active site is precluded by the interaction between Arg353 and Glu248, revealing a mechanism for regulating carboxybiotin access to the BC domain active site. The binding location for the biotin carboxyl carrier protein domain demonstrates that tethered biotin cannot bind in the biotin carboxylase domain active site in the same orientation as free biotin, helping to explain the difference in catalysis observed between tethered biotin and free biotin substrates in biotin carboxylase enzymes. Electron density located in the biotin carboxylase domain active site is assigned to phosphonoacetate, offering a probable location for the putative carboxyphosphate intermediate formed during biotin carboxylation. The insights gained from the T882A Rhizobium etli PC crystal structure provide a new series of catalytic snapshots in PC and offer a revised perspective on catalysis in the biotin-dependent enzyme family.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Bacterial Proteins/genetics , Base Sequence , Biotin/metabolism , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Catalytic Domain , Crystallography, X-Ray , DNA Primers/genetics , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Phosphonoacetic Acid/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Pyruvate Carboxylase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhizobium etli/genetics , Species Specificity , Staphylococcus aureus/enzymology
17.
Biochemistry ; 50(45): 9694-707, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21958066

ABSTRACT

While crystallographic structures of the R. etli pyruvate carboxylase (PC) holoenzyme revealed the location and probable positioning of the essential activator, Mg(2+), and nonessential activator, acetyl-CoA, an understanding of how they affect catalysis remains unclear. The current steady-state kinetic investigation indicates that both acetyl-CoA and Mg(2+) assist in coupling the MgATP-dependent carboxylation of biotin in the biotin carboxylase (BC) domain with pyruvate carboxylation in the carboxyl transferase (CT) domain. Initial velocity plots of free Mg(2+) vs pyruvate were nonlinear at low concentrations of Mg(2+) and a nearly complete loss of coupling between the BC and CT domain reactions was observed in the absence of acetyl-CoA. Increasing concentrations of free Mg(2+) also resulted in a decrease in the K(a) for acetyl-CoA. Acetyl phosphate was determined to be a suitable phosphoryl donor for the catalytic phosphorylation of MgADP, while phosphonoacetate inhibited both the phosphorylation of MgADP by carbamoyl phosphate (K(i) = 0.026 mM) and pyruvate carboxylation (K(i) = 2.5 mM). In conjunction with crystal structures of T882A R. etli PC mutant cocrystallized with phosphonoacetate and MgADP, computational docking studies suggest that phosphonoacetate could coordinate to one of two Mg(2+) metal centers in the BC domain active site. Based on the pH profiles, inhibition studies, and initial velocity patterns, possible mechanisms for the activation, regulation, and coordination of catalysis between the two spatially distinct active sites in pyruvate carboxylase from R. etli by acetyl-CoA and Mg(2+) are described.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Pyruvate Carboxylase/antagonists & inhibitors , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Kinetics , Magnesium/metabolism , Magnesium/pharmacology , Mutagenesis, Site-Directed , Oxaloacetic Acid/metabolism , Phosphonoacetic Acid/pharmacology , Phosphorylation , Protein Structure, Tertiary , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhizobium etli/genetics
18.
Arch Biochem Biophys ; 509(2): 117-26, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21426897

ABSTRACT

2',3'-O-(2,4,6-Trinitrophenyl) adenosine 5'-triphosphate (TNP-ATP) is a fluorescent analogue of ATP. MgTNP-ATP was found to be an allosteric activator of pyruvate carboxylase that exhibits competition with acetyl CoA in activating the enzyme. There is no evidence that MgTNP-ATP binds to the MgATP substrate binding site of the enzyme. At concentrations above saturating, MgATP activates bicarbonate-dependent ATP cleavage, but inhibits the overall reaction. The fluorescence of MgTNP-ATP increases by about 2.5-fold upon binding to the enzyme and decreases on addition of saturating acetyl CoA. However, not all the MgTNP-ATP is displaced by acetyl CoA, or with a combination of saturating concentrations of MgATP and acetyl CoA. The kinetics of the binding of MgTNP-ATP to pyruvate carboxylase have been measured and shown to be triphasic, with the two fastest phases having pseudo first-order rate constants that are dependent on the concentration of MgTNP-ATP. The kinetics of displacement from the enzyme by acetyl CoA have been measured and also shown to be triphasic. A model of the binding process is proposed that links the kinetics of MgTNP-ATP binding to the allosteric activation of the enzyme.


Subject(s)
Acetyl Coenzyme A/metabolism , Adenosine Triphosphate/analogs & derivatives , Fluorescent Dyes/metabolism , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Adenosine Triphosphate/metabolism , Allosteric Regulation , Kinetics , Magnesium Compounds/metabolism , Spectrometry, Fluorescence
20.
Biochemistry ; 49(15): 3296-304, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20230056

ABSTRACT

The roles of Arg548 and Gln552 residues in the active site of the carboxyl transferase domain of Rhizobium etli pyruvate carboxylase were investigated using site-directed mutagenesis. Mutation of Arg548 to alanine or glutamine resulted in the destabilization of the quaternary structure of the enzyme, suggesting that this residue has a structural role. Mutations R548K, Q552N, and Q552A resulted in a loss of the ability to catalyze pyruvate carboxylation, biotin-dependent decarboxylation of oxaloacetate, and the exchange of protons between pyruvate and water. These mutants retained the ability to catalyze reactions that occur at the active site of the biotin carboxylase domain, i.e., bicarbonate-dependent ATP cleavage and ADP phosphorylation by carbamoyl phosphate. The effects of oxamate on the catalysis in the biotin carboxylase domain by the R548K and Q552N mutants were similar to those on the catalysis of reactions by the wild-type enzyme. However, the presence of oxamate had no effect on the reactions catalyzed by the Q552A mutant. We propose that Arg548 and Gln552 facilitate the binding of pyruvate and the subsequent transfer of protons between pyruvate and biotin in the partial reaction catalyzed in the active site of the carboxyl transferase domain of Rhizobium etli pyruvate carboxylase.


Subject(s)
Arginine , Glutamine , Pyruvate Carboxylase/chemistry , Pyruvate Carboxylase/metabolism , Rhizobium etli/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biotin/metabolism , Catalysis , Catalytic Domain , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Multiprotein Complexes/chemistry , Mutagenesis, Site-Directed , Protein Conformation , Protein Structure, Quaternary , Pyruvate Carboxylase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...