Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biochemistry ; 58(14): 1918-1930, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30912442

ABSTRACT

N-Acyl sulfamoyladenosines (acyl-AMS) have been used extensively to inhibit adenylate-forming enzymes that are involved in a wide range of biological processes. These acyl-AMS inhibitors are nonhydrolyzable mimics of the cognate acyl adenylate intermediates that are bound tightly by adenylate-forming enzymes. However, the anionic acyl sulfamate moiety presents a pharmacological liability that may be detrimental to cell permeability and pharmacokinetic profiles. We have previously developed the acyl sulfamate OSB-AMS (1) as a potent inhibitor of the adenylate-forming enzyme MenE, an o-succinylbenzoate-CoA (OSB-CoA) synthetase that is required for bacterial menaquinone biosynthesis. Herein, we report the use of computational docking to develop novel, non-acyl sulfamate inhibitors of MenE. A m-phenyl ether-linked analogue (5) was found to be the most potent inhibitor (IC50 = 8 µM; Kd = 244 nM), and its X-ray co-crystal structure was determined to characterize its binding mode in comparison to the computational prediction. This work provides a framework for the development of potent non-acyl sulfamate inhibitors of other adenylate-forming enzymes in the future.


Subject(s)
Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Succinate-CoA Ligases/antagonists & inhibitors , Vitamin K 2/metabolism , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Kinetics , Microbial Sensitivity Tests , Models, Chemical , Molecular Docking Simulation , Molecular Structure , Mutation , Protein Conformation , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology
2.
Biochemistry ; 54(42): 6514-6524, 2015 10 27.
Article in English | MEDLINE | ID: mdl-26394156

ABSTRACT

MenE is an o-succinylbenzoyl-CoA (OSB-CoA) synthetase in the bacterial menaquinone biosynthesis pathway and is a promising target for the development of novel antibacterial agents. The enzyme catalyzes CoA ligation via an acyl-adenylate intermediate, and we have previously reported tight-binding inhibitors of MenE based on stable acyl-sulfonyladenosine analogues of this intermediate, including OSB-AMS (1), which has an IC50 value of ≤25 nM for Escherichia coli MenE. Herein, we show that OSB-AMS reduces menaquinone levels in Staphylococcus aureus, consistent with its proposed mechanism of action, despite the observation that the antibacterial activity of OSB-AMS is ∼1000-fold lower than the IC50 for enzyme inhibition. To inform the synthesis of MenE inhibitors with improved antibacterial activity, we have undertaken a structure-activity relationship (SAR) study stimulated by the knowledge that OSB-AMS can adopt two isomeric forms in which the OSB side chain exists either as an open-chain keto acid or a cyclic lactol. These studies revealed that negatively charged analogues of the keto acid form bind, while neutral analogues do not, consistent with the hypothesis that the negatively charged keto acid form of OSB-AMS is the active isomer. X-ray crystallography and site-directed mutagenesis confirm the importance of a conserved arginine for binding the OSB carboxylate. Although most lactol isomers tested were inactive, a novel difluoroindanediol inhibitor (11) with improved antibacterial activity was discovered, providing a pathway toward the development of optimized MenE inhibitors in the future.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Succinate-CoA Ligases/antagonists & inhibitors , Succinate-CoA Ligases/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/toxicity , Arginine/chemistry , Catalytic Domain/genetics , Chlorocebus aethiops , Conserved Sequence , Crystallography, X-Ray , Drug Discovery , Escherichia coli K12/enzymology , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenylbutyrates/chemistry , Phenylbutyrates/pharmacology , Phenylbutyrates/toxicity , Protein Conformation , Sequence Homology, Amino Acid , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Structure-Activity Relationship , Succinate-CoA Ligases/genetics , Vero Cells , Vitamin K 2/metabolism
3.
J Inherit Metab Dis ; 37(3): 353-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24154984

ABSTRACT

BACKGROUND: Valproic acid (VPA) is an effective antiepileptic drug that may induce progressive microvesicular steatosis. The impairment of mitochondrial function may be an important metabolic effect of VPA treatment with potential adverse consequences. OBJECTIVE: To investigate the influence of VPA on the activity of GTP- and ATP-specific succinate:CoA ligases (G-SUCL and A-SUCL). METHODS: The GTP- and ATP-specific SUCL activities were measured in human fibroblasts in the reverse direction, i.e. the formation of succinyl-CoA. These were assessed at different concentrations of succinate in the presence of VPA, valproyl-CoA and zinc chloride, an established inhibitor of the enzymes. Activities were measured using an optimized HPLC procedure. RESULTS: Valproyl-CoA (1 mM) inhibited the activity of A-SUCL and G-SUCL by 45-55% and 25-50%, respectively. VPA (1 mM) had no influence on the activity of the two enzymes. DISCUSSION: Valproyl-CoA appears to affect the activity of SUCL, especially with the ATP-specific enzyme. Considering the key role of SUCL in the Krebs cycle, interference with its activity might impair the cellular energy status. Moreover, A-SUCL is bound to the nucleoside diphosphate kinase (NDPK), which is responsible for the mitochondrial (deoxy)nucleotide synthesis. An inhibition of A-SUCL might influence the activity of NDPK inducing an imbalance of nucleotides in the mitochondria and eventually mitochondrial DNA depletion. This may account for the potential liver failure associated with valproate therapy, reported in patients with deficiencies within the mitochondrial DNA replicase system such as polymerase gamma 1.


Subject(s)
Acyl Coenzyme A/pharmacology , Adenosine Triphosphate/physiology , Guanosine Triphosphate/physiology , Succinate-CoA Ligases/antagonists & inhibitors , DNA, Mitochondrial/metabolism , Humans , Liver Failure/chemically induced , Nucleoside-Diphosphate Kinase/physiology , Valproic Acid/adverse effects , Valproic Acid/pharmacology
4.
Chembiochem ; 13(1): 129-36, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22109989

ABSTRACT

MenE, the o-succinylbenzoate (OSB)-CoA synthetase from bacterial menaquinone biosynthesis, is a promising new antibacterial target. Sulfonyladenosine analogues of the cognate reaction intermediate, OSB-AMP, have been developed as inhibitors of the MenE enzymes from Mycobacterium tuberculosis (mtMenE), Staphylococcus aureus (saMenE) and Escherichia coli (ecMenE). Both a free carboxylate and a ketone moiety on the OSB side chain are required for potent inhibitory activity. OSB-AMS (4) is a competitive inhibitor of mtMenE with respect to ATP (K(i) =5.4±0.1 nM) and a noncompetitive inhibitor with respect to OSB (K(i) =11.2±0.9 nM). These data are consistent with a Bi Uni Uni Bi Ping-Pong kinetic mechanism for these enzymes. In addition, OSB-AMS inhibits saMenE with K(i)(app) =22±8 nM and ecMenE with K(i)(OSB) =128±5 nM. Putative active-site residues, Arg222, which may interact with the OSB aromatic carboxylate, and Ser302, which may bind the OSB ketone oxygen, have been identified through computational docking of OSB-AMP with the unliganded crystal structure of saMenE. A pH-dependent interconversion of the free keto acid and lactol forms of the inhibitors is also described, along with implications for inhibitor design.


Subject(s)
Adenosine Monophosphate/pharmacology , Enzyme Inhibitors/pharmacology , Phenylbutyrates/pharmacology , Succinate-CoA Ligases/antagonists & inhibitors , Vitamin K 2/metabolism , Adenosine Monophosphate/chemical synthesis , Adenosine Monophosphate/chemistry , Catalytic Domain/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Escherichia coli/metabolism , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Phenylbutyrates/chemical synthesis , Phenylbutyrates/chemistry , Staphylococcus aureus/enzymology , Staphylococcus aureus/metabolism , Stereoisomerism , Structure-Activity Relationship , Succinate-CoA Ligases/metabolism , Vitamin K 2/chemistry
5.
Biochim Biophys Acta ; 1812(5): 625-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21295139

ABSTRACT

SUCLA2-related mitochondrial DNA (mtDNA) depletion syndrome is a result of mutations in the ß subunit of the ADP-dependent isoform of the Krebs cycle succinyl-CoA synthase (SCS). The mechanism of tissue specificity and mtDNA depletion is elusive but complementation by the GDP-dependent isoform encoded by SUCLG2, and the association with mitochondrial nucleoside diphosphate kinase (NDPK), is a plausible link. We have investigated this relationship by studying SUCLA2 deficient fibroblasts derived from patients and detected normal mtDNA content and normal NDPK activity. However, knockdown of SUCLG2 by shRNA in both patient and control fibroblasts resulted in a significant decrease in mtDNA amount, decreased NDPK and cytochrome c oxidase activities, and a marked growth impairment. This suggests that, SUCLG2, to a higher degree than SUCLA2, is crucial for mtDNA maintenance and that mitochondrial NDPK is involved. Although results pertain to a cell culture system, the findings might explain the pathomechanism and tissue specificity in mtDNA depletion caused by defective SUCLA2.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/genetics , Succinate-CoA Ligases/metabolism , Acyl Coenzyme A/metabolism , Cells, Cultured , Electron Transport Complex IV/metabolism , Fibroblasts/metabolism , Homozygote , Humans , Immunoenzyme Techniques , Mitochondria/metabolism , Mutation/genetics , Nucleoside-Diphosphate Kinase/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Succinate-CoA Ligases/antagonists & inhibitors , Succinate-CoA Ligases/genetics
6.
Biochemistry ; 47(47): 12434-47, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18973344

ABSTRACT

o-Succinylbenzoyl-CoA (OSB-CoA) synthetase (EC 6.2.1.26) catalyzes the ATP-dependent condensation of o-succinylbenzoate (OSB) and CoA to form OSB-CoA, the fourth step of the menaquinone biosynthetic pathway in Bacillus anthracis. Gene knockout studies have highlighted this enzyme as a potential target for the discovery of new antibiotics. Here we report the first studies on the kinetic mechanism of B. anthracis OSB-CoA synthetase, classifying it as an ordered bi uni uni bi ping-pong mechanism. Through a series of pre-steady-state and steady-state kinetic studies in conjunction with direct binding studies, it is demonstrated that CoA, the last substrate to bind, strongly activates the first half-reaction after the first round of turnover. The activation of the first half-reaction is most likely achieved by CoA stabilizing conformations of the enzyme in the "F" form, which slowly isomerize back to the E form. Thus, the kinetic mechanism of OSB-CoA synthetase may be more accurately described as an ordered bi uni uni bi iso ping-pong mechanism. The substrate specificity of OSB-CoA synthetase was probed using a series of OSB analogues with alterations in the carboxylate groups. OSB-CoA shows a strong preference for OSB over all of the analogues tested as none were active except 4-[2-(trifluoromethyl)phenyl]-4-oxobutyric acid which exhibited a 100-fold decrease in k(cat)/K(m). On the basis of an understanding of OSB-CoA synthetase's kinetic mechanism and substrate specificity, a reaction intermediate analogue of OSB-AMP, 5'-O-{N-[2-(trifluoromethyl)phenyl]-4-oxobutyl}adenosine sulfonamide (TFMP-butyl-AMS), was designed and synthesized. This inhibitor was found to be an uncompetitive inhibitor to CoA and a mixed-type inhibitor to ATP and OSB with low micromolar inhibition constants. Collectively, these results should serve as an important forerunner to more detailed and extensive inhibitor design studies aimed at developing lead compounds against the OSB-CoA synthetase class of enzymes.


Subject(s)
Bacillus anthracis/enzymology , Biomimetic Materials/pharmacology , Enzyme Inhibitors/pharmacology , Succinate-CoA Ligases/antagonists & inhibitors , Succinate-CoA Ligases/metabolism , Adenosine Triphosphate/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Coenzyme A/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Kinetics , Phenols/metabolism , Phenylbutyrates/metabolism , Substrate Specificity
7.
Cell Metab ; 5(4): 253-64, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17403370

ABSTRACT

Nucleotide-specific isoforms of the tricarboxylic acid (TCA) cycle enzyme succinyl-CoA synthetase (SCS) catalyze substrate-level synthesis of mitochondrial GTP (mtGTP) and ATP (mtATP). While mtATP yield from glucose metabolism is coupled with oxidative phosphorylation and can vary, each molecule of glucose metabolized within pancreatic beta cells produces approximately one mtGTP, making mtGTP a potentially important fuel signal. In INS-1 832/13 cells and cultured rat islets, siRNA suppression of the GTP-producing pathway (DeltaSCS-GTP) reduced glucose-stimulated insulin secretion (GSIS) by 50%, while suppression of the ATP-producing isoform (DeltaSCS-ATP) increased GSIS 2-fold. Insulin secretion correlated with increases in cytosolic calcium, but not with changes in NAD(P)H or the ATP/ADP ratio. These data suggest a role for mtGTP in controlling pancreatic GSIS through modulation of mitochondrial metabolism, possibly involving mitochondrial calcium. Furthermore, in light of its tight coupling to TCA oxidation rates, mtGTP production may serve as an important molecular signal of TCA-cycle activity.


Subject(s)
Glucose/pharmacology , Guanosine Triphosphate/physiology , Insulin/metabolism , Mitochondria/drug effects , Animals , Calcium/metabolism , Cells, Cultured , Energy Metabolism/physiology , Guanosine Triphosphate/analysis , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mitochondria/chemistry , Models, Biological , Oxidation-Reduction , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Succinate-CoA Ligases/antagonists & inhibitors , Succinate-CoA Ligases/genetics
8.
Biochemistry ; 38(22): 7273-83, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10353839

ABSTRACT

Succinyl-CoA synthetase (SCS) catalyzes the reversible interchange of purine nucleoside diphosphate, succinyl-CoA, and Pi with purine nucleoside triphosphate, succinate, and CoA via a phosphorylated histidine (H246alpha) intermediate. Two potential nucleotide-binding sites were predicted in the beta-subunit, and have been differentiated by photoaffinity labeling with 8-N3-ATP and by site-directed mutagenesis. It was demonstrated that 8-N3-ATP is a suitable analogue for probing the nucleotide-binding site of SCS. Two tryptic peptides from the N-terminal domain of the beta-subunit were labeled with 8-N3-ATP. These corresponded to residues 107-119beta and 121-146beta, two regions lying along one side of an ATP-grasp fold. A mutant protein with changes on the opposite side of the fold (G53betaV/R54betaE) was unable to be phosphorylated using ATP or GTP, but could be phosphorylated by succinyl-CoA and Pi. A mutant protein designed to probe nucleotide specificity (P20betaQ) had a Km(app) for GTP that was more than 5 times lower than that of wild-type SCS, whereas parameters for the other substrates remained unchanged. Mutations of residues in the C-terminal domain of the beta-subunit designed to distrupt one loop of the Rossmann fold (I322betaA, and R324betaN/D326betaA) had the greatest effect on the binding of succinate and CoA. They did not disrupt the phosphorylation of SCS with nucleotides. It was concluded that the nucleotide-binding site is located in the N-terminal domain of the beta-subunit. This implies that there are two active sites approximately 35 A apart, and that the H246alpha loop moves between them during catalysis.


Subject(s)
Escherichia coli/enzymology , Purine Nucleotides/chemistry , Purine Nucleotides/metabolism , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Azides/chemistry , Azides/metabolism , Binding Sites , Conserved Sequence , Enzyme Activation , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Phosphorylation , Photoaffinity Labels/chemistry , Photoaffinity Labels/metabolism , Substrate Specificity , Succinate-CoA Ligases/antagonists & inhibitors , Succinate-CoA Ligases/genetics
9.
Mol Biochem Parasitol ; 100(1): 53-9, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10376993

ABSTRACT

Recent drug screenings for new antibacterial drugs directed against histidine phospho-relay signalling pathways in bacteria have resulted in compounds which potently inhibit the histidine kinase activity of bacterial two-component systems. The present study demonstrates that one of these compounds, LY266500, is also a potent inhibitor of histidine phosphorylation in the unicellular eukaryotic parasite Trypanosoma brucei, both in vitro and in whole cells. In vitro, it inhibits histidine phosphorylation of mitochondrial succinyl CoA synthetase. LY26650 does not interfere with the phosphotransfer from the histidine-phosphorylated protein to ADP. In standardized cell culture tests, LY266500 potently inhibits the proliferation of the human pathogens T. brucei rhodesiense and Leishmania donovani. Since the inhibitory activity in vivo is life-cycle stage specific and correlates well with the mitochondrial activity in the different stages, the effect of LY266500 is most likely due to its specific inhibition of the mitochondrial succinyl CoA synthetase.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Kinases , Succinate-CoA Ligases/antagonists & inhibitors , Thiazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/enzymology , Animals , Histidine/metabolism , Histidine Kinase , Humans , Leishmania donovani/drug effects , Mitochondria/enzymology , Phosphorylation , Protein Kinase Inhibitors , Signal Transduction/drug effects , Succinate-CoA Ligases/metabolism , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei rhodesiense/drug effects
10.
Arch Biochem Biophys ; 347(1): 103-12, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9344470

ABSTRACT

Concentrations of GDP, which are expected to bind to the catalytic site and inhibit the autophosphorylation of succinyl-CoA synthetase (SCS) when NTP is used as a substrate, were found to increase the level of phosphoenzyme formed. The ability of GDP to do so is dependent upon the presence of a protein distinct from SCS. The effector protein could be separated from SCS by ammonium sulfate fractionation. Reconstitution experiments show that the protein inhibits SCS, that the inhibition is relieved by GDP, and that the inhibitor recognizes both Escherichia coli and eukaryotic forms of SCS. The inhibitor is itself regulated by the conditions used to grow the bacteria and in a manner that appears distinct from that of SCS.


Subject(s)
Bacterial Proteins/isolation & purification , Enzyme Inhibitors/isolation & purification , Escherichia coli Proteins , Escherichia coli/enzymology , Guanosine Diphosphate/metabolism , RNA-Binding Proteins , Succinate-CoA Ligases/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Allosteric Site , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Binding Sites , Dictyostelium/enzymology , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/pharmacology , Phosphorylation , Protein Denaturation , Succinate-CoA Ligases/antagonists & inhibitors , Temperature
11.
J Bacteriol ; 179(19): 6061-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9324253

ABSTRACT

o-Succinylbenzoyl coenzyme A (OSB-CoA) synthetase, when treated with diethylpyrocarbonate (DEP), showed a time-dependent loss of enzyme activity. The inactivation follows pseudo-first-order kinetics with a second-order rate constant of 9.2 x 10(-4) +/- 1.4 x 10(-4) microM(-1) min(-1). The difference spectrum of the modified enzyme versus the native enzyme showed an increase in A242 that is characteristic of N-carbethoxyhistidine and was reversed by treatment with hydroxylamine. Inactivation due to nonspecific secondary structural changes in the protein and modification of tyrosine, lysine, or cysteine residues was ruled out. Kinetics of enzyme inactivation and the stoichiometry of histidine modification indicate that of the eight histidine residues modified per subunit of the enzyme, a single residue is responsible for the enzyme activity. A plot of the log reciprocal of the half-time of inactivation against the log DEP concentration further suggests that one histidine residue is involved in the catalysis. Further, the enzyme was partially protected from inactivation by either o-succinylbenzoic acid (OSB), ATP, or ATP plus Mg2+ while inactivation was completely prevented by the presence of the combination of OSB, ATP, and Mg2+. Thus, it appears that a histidine residue located at or near the active site of the enzyme is essential for activity. When His341 present in the previously identified ATP binding motif was mutated to Ala, the enzyme lost 65% of its activity and the Km for ATP increased 5.4-fold. Thus, His341 of OSB-CoA synthetase plays an important role in catalysis since it is probably involved in the binding of ATP to the enzyme.


Subject(s)
Escherichia coli/metabolism , Histidine/chemistry , Succinate-CoA Ligases/chemistry , Succinate-CoA Ligases/metabolism , Vitamin K/biosynthesis , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Binding Sites , Diethyl Pyrocarbonate/pharmacology , Hydroxylamine , Hydroxylamines/pharmacology , Kinetics , Magnesium/pharmacology , Mutagenesis, Site-Directed , Phenylbutyrates/pharmacology , Spectrophotometry, Ultraviolet , Succinate-CoA Ligases/antagonists & inhibitors
13.
J Enzyme Inhib ; 10(2): 141-6, 1996.
Article in English | MEDLINE | ID: mdl-8835939

ABSTRACT

The effect of uroporphyrin I (UI) on several cytosolic and mitochondrial enzymes (succinyl CoA synthetase, delta-aminolevulinic acid synthetase, rhodanese, lactate dehydrogenase) has been examined. All the enzymes were inactivated in the presence of the porphyrin both in the dark and under UV light.


Subject(s)
L-Lactate Dehydrogenase/antagonists & inhibitors , Thiosulfate Sulfurtransferase/antagonists & inhibitors , Uroporphyrins/pharmacology , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Animals , Cattle , Cytoplasm/enzymology , Enzyme Inhibitors/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Mitochondria, Liver/enzymology , Mitochondria, Muscle/enzymology , Photosensitizing Agents/pharmacology , Rabbits , Succinate-CoA Ligases/antagonists & inhibitors , Ultraviolet Rays/adverse effects
15.
Gen Physiol Biophys ; 10(1): 71-82, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1869044

ABSTRACT

Effect of vanadate and vanadyl ions on the ATP-dependent succinyl-CoA synthetase (A-SCS) solubilized by Lubrol-PX from the rat brain mitochondria was tested. Vanadate added to the assay medium at 10(-5) mol.l-1 and 10(-4) mol.l-1 concentrations inhibited the enzyme activity by about 50% and 94%, respectively. When the enzyme was solubilized from the mitochondria preincubated with 10(-4) mol.l-1 and 10(-3) mol.l-1 vanadate, the residual inhibitions were 55% and 100% respectively. The vanadyl cation also induced inhibition of the A-SCS activity but the effect was less expressed. At 10(-4) mol.l-1 concentration only 20% inhibition was achieved. The A-SCS solubilized from the mitochondrial subfractions (perikaryal, light and heavy synaptosomal) differed neither in the activity of A-SCS nor in the susceptibility toward action of vanadium ions. A strong dependence of the vanadate inhibition on the concentration of succinate was observed. The above effect (50% inhibition) could be demonstrated only at saturating concentration of succinate (50 mmol.l-1). The mechanism of vanadium ions action as well as differences between vanadate and vanadyl ions effects are discussed.


Subject(s)
Succinate-CoA Ligases/antagonists & inhibitors , Vanadates/pharmacology , Animals , Dose-Response Relationship, Drug , Kinetics , Mitochondria/drug effects , Mitochondria/enzymology , Neurons/cytology , Rats , Synaptosomes/drug effects , Synaptosomes/enzymology
16.
Biochem Cell Biol ; 65(5): 452-7, 1987 May.
Article in English | MEDLINE | ID: mdl-3304349

ABSTRACT

Succinyl-CoA synthetase catalyzes the substrate-level phosphorylation step of the tricarboxylic acid cycle. The enzyme, as isolated from Escherichia coli, has an alpha 2 beta 2 subunit structure. It is known that substrate-binding sites are distributed between both subunit types and that the active enzyme is the nondissociating tetramer. This paper describes a study of the process of assembly of the enzyme from its denatured constituent subunits. Starting with equimolar mixtures of the subunits that are prepared in denaturing conditions (6 M urea, 5% acetic acid), rapid renaturation to produce virtually a fully active enzyme occurs after neutralization and dilution under suitable conditions. This process occurs most efficiently in the presence of either ATP or Pi, indicating that occupation of the phosphoryl-binding site on the refolding alpha subunit facilitates productive intrasubunit interactions. We have determined conditions of protein concentration, pH, temperature, final urea concentration, and buffer compositions that optimize both the rate and extent of production of active enzyme. The final refolded product is indistinguishable from the native species with respect to its specific catalytic activity, size, and other physical properties. To probe further the mechanism and route of renaturation, we have shown that the rate of appearance of activity has first-order dependence on each of the two subunits. The step that determines the rate of assembly is thus bimolecular, such as the association of structural monomers to form a dimeric transient species. The highly specific mutual interactions between the refolding transient species of subunits must be essential for the correct assembly of this enzyme from the two gene products in vivo.


Subject(s)
Coenzyme A Ligases/analysis , Escherichia coli/enzymology , Succinate-CoA Ligases/analysis , Adenosine Triphosphate , Chromatography, Gel , Hydrogen-Ion Concentration , Kinetics , Macromolecular Substances , Magnesium , Magnesium Chloride , Phosphates , Succinate-CoA Ligases/antagonists & inhibitors , Temperature , Urea/pharmacology
17.
FEBS Lett ; 196(2): 341-3, 1986 Feb 17.
Article in English | MEDLINE | ID: mdl-2936621

ABSTRACT

The activity of succinyl-CoA synthetase from mouse liver and kidney was inhibited by streptozotocin in vitro. Streptozotocin behaved essentially as a non-competitive inhibitor, and the following kinetic values were obtained (in the presence of 10 nM streptozotocin): apparent Km 1.7 mM, apparent Ki 10 nM, and kcat 440 nkat X kg-1. Compared with non-diabetic control mice, the succinyl-CoA synthetase activity was significantly decreased in the islets and kidneys of mice with early (1 h) and manifest (greater than or equal to 2 days) streptozotocin diabetes, whereas the activity in the liver was not significantly altered. Inhibited succinyl-CoA synthetase activity is believed to play a prominent role in the cellular effects of streptozotocin.


Subject(s)
Coenzyme A Ligases/antagonists & inhibitors , Streptozocin/pharmacology , Succinate-CoA Ligases/antagonists & inhibitors , Animals , Diabetes Mellitus, Experimental/enzymology , Female , In Vitro Techniques , Kidney/enzymology , Kinetics , Liver/enzymology , Male , Mice , Mice, Inbred C57BL
18.
FEBS Lett ; 178(2): 245-8, 1984 Dec 10.
Article in English | MEDLINE | ID: mdl-6510522

ABSTRACT

Considerable variations were found in the in vitro effect of alloxan on mouse liver enzymes associated with the citric acid cycle. The following approximative alloxan concentrations induced 50% inhibition of enzyme activity: 10(-6)M for aconitase, 10(-4)M for NAD-linked isocitrate dehydrogenase, glutamate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinyl-CoA synthetase and fumarase, and 10(-3)M for citrate synthase and NADP-linked isocitrate dehydrogenase. Pyruvate dehydrogenase, succinate dehydrogenase and malate dehydrogenase were not inhibited by 10(-3)M alloxan. The inhibition of aconitase was competitive both when using mouse liver and purified porcine heart enzyme. The Ki values for the purified enzyme in the presence of 5 microM alloxan were 0.22 microM with citrate, 4.0 microM with cis-aconitate and 0.62 microM with isocitrate as substrate. The high sensitivity of aconitase for inhibition by alloxan probably plays a prominent role for the toxic effects of alloxan.


Subject(s)
Aconitate Hydratase/antagonists & inhibitors , Alloxan/pharmacology , Citric Acid Cycle , Mitochondria, Liver/enzymology , Animals , Citrate (si)-Synthase/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Glutamate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Malate Dehydrogenase/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Succinate-CoA Ligases/antagonists & inhibitors
19.
J Biol Chem ; 259(15): 9642-5, 1984 Aug 10.
Article in English | MEDLINE | ID: mdl-6378911

ABSTRACT

Adenosine 5'-O-(3-thio)triphosphate (ATP gamma S) has been shown to be a potent inhibitor of Escherichia coli succinyl-CoA synthetase. This inhibition was competitive with respect to ATP and GTP (Ki values of 0.8 and 0.7 microM, respectively) and mixed with respect to CoA and succinate. ATP gamma S previously had been shown to be a weak substrate of the enzyme, probably because of the relatively sluggish reactivity of the thiophosphoryl enzyme intermediate (Wolodko, W. T., Brownie, E. R., O'Connor, M. D., and Bridger, W. A. (1983) J. Biol. Chem. 258, 14116-14119). In our work, reaction of thiophosphoryl enzyme with ADP was greatly stimulated by succinyl-CoA, an observation that is consistent with the concept of alternating-sites cooperativity. Thiophosphoryl group release did not appear to be accompanied by "other-site" phosphorylation, in contrast to ATP stimulation of thiophosphoryl group release in the presence of succinate and CoA (Wolodko et al., see above). In addition, ADP did not appear to be required in the latter reaction.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Affinity Labels/metabolism , Coenzyme A Ligases/antagonists & inhibitors , Escherichia coli/enzymology , Succinate-CoA Ligases/antagonists & inhibitors , Thionucleotides/metabolism , Acyl Coenzyme A/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Binding, Competitive , Coenzyme A/metabolism , Succinates/metabolism , Succinic Acid
20.
Biochem J ; 215(3): 513-8, 1983 Dec 01.
Article in English | MEDLINE | ID: mdl-6362660

ABSTRACT

Escherichia coli succinyl-CoA synthetase (EC 6.2.1.5) was irreversibly inactivated on incubation with the adenine nucleotide analogue 5'-p-fluorosulphonylbenzoyladenosine (5'-FSBA). Optimal inactivation by 5'-FSBA took place in 40% (v/v) dimethylformamide. ATP and ADP protected the enzyme against inactivation by 5'-FSBA, whereas desulpho-CoA, an analogue of CoA, did not. Inactivation of succinyl-CoA synthetase by 5'-FSBA resulted in total loss of almost four thiol groups per alpha beta-dimer, of which two groups appeared to be essential for catalytic activity. 5'-FSBA at the first instance appeared to interact non-specifically with non-essential thiol groups, followed by a more specific reaction with essential thiol groups in the ATP(ADP)-binding region. Plots of the data according to the method of Tsou [(1962) Sci. Sin. 11, 1535-1558] revealed that, of the two slower-reacting thiol groups, only one was essential for catalytic activity. When succinyl-CoA synthetase that had been totally inactivated by 5'-FSBA was unfolded in acidic urea and then refolded in the presence of 100 mM-dithiothreitol, 85% of the activity, in comparison with the appropriate control, was restored. These data are interpreted to indicate that inactivation of succinyl-CoA synthetase by 5'-FSBA involves the formation of a disulphide bond between two cysteine residues. Disulphide bond formation likely proceeds via a thiosulphonate intermediate between 5'-p-sulphonylbenzoyladenosine and one of the reactive thiol groups of the enzyme.


Subject(s)
Adenosine/analogs & derivatives , Coenzyme A Ligases/antagonists & inhibitors , Escherichia coli/enzymology , Succinate-CoA Ligases/antagonists & inhibitors , Adenosine/pharmacology , Binding Sites , Dithiothreitol/pharmacology , Ligands , Protein Conformation , Sulfhydryl Compounds
SELECTION OF CITATIONS
SEARCH DETAIL
...