Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Biochemistry (Mosc) ; 88(5): 600-609, 2023 May.
Article in English | MEDLINE | ID: mdl-37331706

ABSTRACT

O-acetylhomoserine sulfhydrylase is one of the key enzymes in biosynthesis of methionine in Clostridioides difficile. The mechanism of γ-substitution reaction of O-acetyl-L-homoserine catalyzed by this enzyme is the least studied among the pyridoxal-5'-phosphate-dependent enzymes involved in metabolism of cysteine and methionine. To clarify the role of active site residues Tyr52 and Tyr107, four mutant forms of the enzyme with replacements of these residues with phenylalanine and alanine were generated. Catalytic and spectral properties of the mutant forms were investigated. The rate of γ-substitution reaction catalyzed by the mutant forms with replaced Tyr52 residue decreased by more than three orders of magnitude compared to the wild-type enzyme. The Tyr107Phe and Tyr107Ala mutant forms practically did not catalyze this reaction. Replacements of the Tyr52 and Tyr107 residues led to the decrease in affinity of apoenzyme to coenzyme by three orders of magnitude and changes in the ionic state of the internal aldimine of the enzyme. The obtained results allowed us to assume that Tyr52 is involved in ensuring optimal position of the catalytic coenzyme-binding lysine residue at the stages of C-α-proton elimination and elimination of the side group of the substrate. Tyr107 could act as a general acid catalyst at the stage of acetate elimination.


Subject(s)
Clostridioides difficile , Clostridioides difficile/metabolism , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Catalytic Domain , Clostridioides/metabolism , Tyrosine , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Methionine , Kinetics
2.
Mol Cell Proteomics ; 20: 100098, 2021.
Article in English | MEDLINE | ID: mdl-34022432

ABSTRACT

CysE and CysK, the last two enzymes of the cysteine biosynthetic pathway, engage in a bienzyme complex, cysteine synthase, with yet incompletely characterized three-dimensional structure and regulatory function. Being absent in mammals, the two enzymes and their complex are attractive targets for antibacterial drugs. We have used hydrogen/deuterium exchange MS to unveil how complex formation affects the conformational dynamics of CysK and CysE. Our results support a model where CysE is present in solution as a dimer of trimers, and each trimer can bind one CysK homodimer. When CysK binds to one CysE monomer, intratrimer allosteric communication ensures conformational and dynamic symmetry within the trimer. Furthermore, a long-range allosteric signal propagates through CysE to induce stabilization of the interface between the two CysE trimers, preparing the second trimer for binding the second CysK with a nonrandom orientation. These results provide new molecular insights into the allosteric formation of the cysteine synthase complex and could help guide antibacterial drug design.


Subject(s)
Cysteine Synthase/chemistry , Chromatography, Liquid , Deuterium , Deuterium Exchange Measurement , Hydrogen , Mass Spectrometry
3.
J Biol Chem ; 296: 100041, 2021.
Article in English | MEDLINE | ID: mdl-33162395

ABSTRACT

O-acetyl serine sulfhydrylase (OASS), referred to as cysteine synthase (CS), synthesizes cysteine from O-acetyl serine (OAS) and sulfur in bacteria and plants. The inherent challenge for CS is to overcome 4 to 6 log-folds stronger affinity for its natural inhibitor, serine acetyltransferase (SAT), as compared with its affinity for substrate, OAS. Our recent study showed that CS employs a novel competitive-allosteric mechanism to selectively recruit its substrate in the presence of natural inhibitor. In this study, we trace the molecular features that control selective substrate recruitment. To generalize our findings, we used CS from three different bacteria (Haemophilus, Salmonella, and Mycobacterium) as our model systems and analyzed structural and substrate-binding features of wild-type CS and its ∼13 mutants. Results show that CS uses a noncatalytic residue, M120, located 20 Šaway from the reaction center, to discriminate in favor of substrate. M120A and background mutants display significantly reduced substrate binding, catalytic efficiency, and inhibitor binding. Results shows that M120 favors the substrate binding by selectively enhancing the affinity for the substrate and disengaging the inhibitor by 20 to 286 and 5- to 3-folds, respectively. Together, M120 confers a net discriminative force in favor of substrate by 100- to 858-folds.


Subject(s)
Cysteine Synthase/metabolism , Allosteric Regulation , Amino Acid Sequence , Amino Acid Substitution , Catalysis , Circular Dichroism , Crystallography, X-Ray , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Methionine/chemistry , Protein Conformation , Substrate Specificity
4.
Biochemistry ; 60(7): 524-536, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33539704

ABSTRACT

Serine synthase (SS) from Fusobacterium nucleatum is a fold type II pyridoxal 5'-phosphate (PLP)-dependent enzyme that catalyzes the ß-replacement of l-cysteine with water to form l-serine and H2S. Herein, we show that SS can also function as a cysteine synthase, catalyzing the ß-replacement of l-serine with bisulfide to produce l-cysteine and H2O. The forward (serine synthase) and reverse (cysteine synthase) reactions occur with comparable turnover numbers and catalytic efficiencies for the amino acid substrate. Reaction of SS with l-cysteine leads to transient formation of a quinonoid species, suggesting that deprotonation of the Cα and ß-elimination of the thiolate group from l-cysteine occur via a stepwise mechanism. In contrast, the quinonoid species was not detected in the formation of the α-aminoacrylate intermediate following reaction of SS with l-serine. A key active site residue, D232, was shown to stabilize the more chemically reactive ketoenamine PLP tautomer and also function as an acid/base catalyst in the forward and reverse reactions. Fluorescence resonance energy transfer between PLP and W99, the enzyme's only tryptophan residue, supports ligand-induced closure of the active site, which shields the PLP cofactor from the solvent and increases the basicity of D232. These results provide new insight into amino acid metabolism in F. nucleatum and highlight the multiple catalytic roles of D232 in a new member of the fold type II family of PLP-dependent enzymes.


Subject(s)
Cysteine Synthase/metabolism , Fusobacterium nucleatum/metabolism , Alanine/analogs & derivatives , Binding Sites , Catalysis , Catalytic Domain , Cysteine/chemistry , Cysteine Synthase/chemistry , Fusobacterium nucleatum/enzymology , Kinetics , Ligands , Models, Molecular , Protein Conformation , Pyridoxal Phosphate/metabolism , Serine/chemistry
5.
Biochem J ; 475(4): 733-748, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29343611

ABSTRACT

Hydrogen sulfide (H2S) plays important roles in the pathogenesis of periodontitis. Oral pathogens typically produce H2S from l-cysteine in addition to pyruvate and [Formula: see text] However, fn1055 from Fusobacterium nucleatum subsp. nucleatum ATCC 25586 encodes a pyridoxal 5'-phosphate (PLP)-dependent enzyme that catalyzes the production of H2S and l-serine from l-cysteine and H2O, an unusual cysteine (hydroxyl) lyase reaction (ß-replacement reaction). To reveal the reaction mechanism, the crystal structure of substrate-free Fn1055 was determined. Based on this structure, a model of the l-cysteine-PLP Schiff base suggested that the thiol group forms hydrogen bonds with Asp232 and Ser74, and the substrate α-carboxylate interacts with Thr73 and Gln147 Asp232 is a unique residue to Fn1055 and its substitution to asparagine (D232N) resulted in almost complete loss of ß-replacement activity. The D232N structure obtained in the presence of l-cysteine contained the α-aminoacrylate-PLP Schiff base in the active site, indicating that Asp232 is essential for the addition of water to the α-aminoacrylate to produce the l-serine-PLP Schiff base. Rapid-scan stopped-flow kinetic analyses showed an accumulation of the α-aminoacrylate intermediate during the reaction cycle, suggesting that water addition mediated by Asp232 is the rate-limiting step. In contrast, mutants containing substitutions of other active-site residues (Ser74, Thr73, and Gln147) exhibited reduced ß-replacement activity by more than 100-fold. Finally, based on the structural and biochemical analyses, we propose a mechanism of the cysteine (hydroxyl) lyase reaction by Fn1055. The present study leads to elucidation of the H2S-producing mechanism in F. nucleatum.


Subject(s)
Cysteine Synthase/chemistry , Cysteine/chemistry , Fusobacterium nucleatum/enzymology , Protein Conformation , Catalysis , Catalytic Domain , Crystallography, X-Ray , Cysteine/metabolism , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Fusobacterium nucleatum/pathogenicity , Humans , Hydrogen Sulfide/chemistry , Hydrogen Sulfide/metabolism , Hydroxyl Radical/chemistry , Kinetics , Models, Molecular , Schiff Bases/chemistry
6.
Proc Natl Acad Sci U S A ; 113(35): 9792-7, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27531961

ABSTRACT

Contact-dependent growth inhibition (CDI) is a widespread mechanism of bacterial competition. CDI(+) bacteria deliver the toxic C-terminal region of contact-dependent inhibition A proteins (CdiA-CT) into neighboring target bacteria and produce CDI immunity proteins (CdiI) to protect against self-inhibition. The CdiA-CT(EC536) deployed by uropathogenic Escherichia coli 536 (EC536) is a bacterial toxin 28 (Ntox28) domain that only exhibits ribonuclease activity when bound to the cysteine biosynthetic enzyme O-acetylserine sulfhydrylase A (CysK). Here, we present crystal structures of the CysK/CdiA-CT(EC536) binary complex and the neutralized ternary complex of CysK/CdiA-CT/CdiI(EC536) CdiA-CT(EC536) inserts its C-terminal Gly-Tyr-Gly-Ile peptide tail into the active-site cleft of CysK to anchor the interaction. Remarkably, E. coli serine O-acetyltransferase uses a similar Gly-Asp-Gly-Ile motif to form the "cysteine synthase" complex with CysK. The cysteine synthase complex is found throughout bacteria, protozoa, and plants, indicating that CdiA-CT(EC536) exploits a highly conserved protein-protein interaction to promote its toxicity. CysK significantly increases CdiA-CT(EC536) thermostability and is required for toxin interaction with tRNA substrates. These observations suggest that CysK stabilizes the toxin fold, thereby organizing the nuclease active site for substrate recognition and catalysis. By contrast, Ntox28 domains from Gram-positive bacteria lack C-terminal Gly-Tyr-Gly-Ile motifs, suggesting that they do not interact with CysK. We show that the Ntox28 domain from Ruminococcus lactaris is significantly more thermostable than CdiA-CT(EC536), and its intrinsic tRNA-binding properties support CysK-independent nuclease activity. The striking differences between related Ntox28 domains suggest that CDI toxins may be under evolutionary pressure to maintain low global stability.


Subject(s)
Bacterial Toxins/chemistry , Contact Inhibition/genetics , Cysteine Synthase/chemistry , Escherichia coli Proteins/chemistry , Uropathogenic Escherichia coli/chemistry , Amino Acid Sequence , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Secondary , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ruminococcus/chemistry , Ruminococcus/metabolism , Substrate Specificity , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/metabolism
7.
Int J Mol Sci ; 20(20)2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31640223

ABSTRACT

The formation of multienzymatic complexes allows for the fine tuning of many aspects of enzymatic functions, such as efficiency, localization, stability, and moonlighting. Here, we investigated, in solution, the structure of bacterial cysteine synthase (CS) complex. CS is formed by serine acetyltransferase (CysE) and O-acetylserine sulfhydrylase isozyme A (CysK), the enzymes that catalyze the last two steps of cysteine biosynthesis in bacteria. CysK and CysE have been proposed as potential targets for antibiotics, since cysteine and related metabolites are intimately linked to protection of bacterial cells against redox damage and to antibiotic resistance. We applied a combined approach of small-angle X-ray scattering (SAXS) spectroscopy and protein painting to obtain a model for the solution structure of CS. Protein painting allowed the identification of protein-protein interaction hotspots that were then used as constrains to model the CS quaternary assembly inside the SAXS envelope. We demonstrate that the active site entrance of CysK is involved in complex formation, as suggested by site-directed mutagenesis and functional studies. Furthermore, complex formation involves a conformational change in one CysK subunit that is likely transmitted through the dimer interface to the other subunit, with a regulatory effect. Finally, SAXS data indicate that only one active site of CysK is involved in direct interaction with CysE and unambiguously unveil the quaternary arrangement of CS.


Subject(s)
Bacteria/enzymology , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Serine O-Acetyltransferase/chemistry , Serine O-Acetyltransferase/metabolism , Bacteria/chemistry , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cysteine Synthase/genetics , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Protein Interaction Maps , Scattering, Small Angle , Serine O-Acetyltransferase/genetics , X-Ray Diffraction
8.
J Chem Inf Model ; 58(3): 710-723, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29481752

ABSTRACT

Saturation transfer difference (STD) is an NMR technique conventionally applied in drug discovery to identify ligand moieties relevant for binding to protein cavities. This is important to direct medicinal chemistry efforts in small-molecule optimization processes. However, STD does not provide any structural details about the ligand-target complex under investigation. Herein, we report the application of a new integrated approach, which combines enhanced sampling methods with STD experiments, for the characterization of ligand-target complexes that are instrumental for drug design purposes. As an example, we have studied the interaction between StOASS-A, a potential antibacterial target, and an inhibitor previously reported. This approach allowed us to consider the ligand-target complex from a dynamic point of view, revealing the presence of an accessory subpocket which can be exploited to design novel StOASS-A inhibitors. As a proof of concept, a small library of derivatives was designed and evaluated in vitro, displaying the expected activity.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Salmonella typhimurium/enzymology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Cysteine Synthase/chemistry , Drug Design , Ligands , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation , Molecular Dynamics Simulation , Salmonella typhimurium/drug effects , Thermodynamics
9.
Biochem J ; 474(7): 1221-1239, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28126739

ABSTRACT

Cysteine biosynthesis takes place via a two-step pathway in bacteria, fungi, plants and protozoan parasites, but not in humans, and hence, the machinery of cysteine biosynthesis is an opportune target for therapeutics. The decameric cysteine synthase complex (CSC) is formed when the C-terminal tail of serine acetyltransferase (SAT) binds in the active site of O-acetylserine sulfydrylase (OASS), playing a role in the regulation of this pathway. Here, we show that OASS from Brucella abortus (BaOASS) does not interact with its cognate SAT C-terminal tail. Crystal structures of native BaOASS showed that residues Gln96 and Tyr125 occupy the active-site pocket and interfere with the entry of the SAT C-terminal tail. The BaOASS (Q96A-Y125A) mutant showed relatively strong binding (Kd = 32.4 µM) to BaSAT C-terminal peptides in comparison with native BaOASS. The mutant structure looks similar except that the active-site pocket has enough space to bind the SAT C-terminal end. Surface plasmon resonance results showed a relatively strong (7.3 µM Kd) interaction between BaSAT and the BaOASS (Q96A-Y125A), but no interaction with native BaOASS. Taken together, our observations suggest that the CSC does not form in B. abortus.


Subject(s)
Bacterial Proteins/chemistry , Brucella abortus/chemistry , Cysteine Synthase/chemistry , Cysteine/biosynthesis , Serine O-Acetyltransferase/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucella abortus/enzymology , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine O-Acetyltransferase/genetics , Serine O-Acetyltransferase/metabolism , Structure-Activity Relationship , Substrate Specificity
10.
J Plant Res ; 131(2): 319-329, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29181648

ABSTRACT

In the cysteine and mimosine biosynthesis process, O-acetyl-L-serine (OAS) is the common substrate. In the presence of O-acetylserine (thiol) lyase (OASTL, cysteine synthase) the reaction of OAS with sulfide produces cysteine, while with 3-hydroxy-4-pyridone (3H4P) produces mimosine. The enzyme OASTL can either catalyze Cys synthesis or both Cys and mimosine. A cDNA for cytosolic OASTL was cloned from M. pudica for the first time containing 1,410 bp nucleotides. The purified protein product from overexpressed bacterial cells produced Cys only, but not mimosine, indicating it is Cys specific. Kinetic studies revealed that pH and temperature optima for Cys production were 6.5 and 50 °C, respectively. The measured Km, Kcat, and Kcat Km-1 values were 159 ± 21 µM, 33.56 s-1, and 211.07 mM-1s-1 for OAS and 252 ± 25 µM, 32.99 s-1, and 130.91 mM-1s-1 for Na2S according to the in vitro Cys assay. The Cy-OASTL of Mimosa pudica is specific to Cys production, although it contains sensory roles in sulfur assimilation and the reduction network in the intracellular environment of M. pudica.


Subject(s)
Cysteine Synthase/genetics , Mimosa/genetics , Mimosine/metabolism , Plant Proteins/genetics , Amino Acid Sequence , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Cytosol/metabolism , Mimosa/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Sequence Alignment
11.
Biochemistry ; 56(18): 2385-2399, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28414426

ABSTRACT

Serine acetyltransferase (SAT) and O-acetylserine sulfhydrylase (OASS), which catalyze the last two steps of cysteine biosynthesis, interact and form the cysteine regulatory complex (CRC). The current model of Salmonella typhimurium predicts that CRC is composed of one [SAT]hexamer unit and two molecules of [OASS]dimer. However, it is not clear why [SAT]hexamer cannot engage all of its six high-affinity binding sites. We examined the assembly state(s) of CRC by size exclusion chromatography, analytical ultracentrifugation (AUC), isothermal titration calorimetry (ITC), and surface plasmon resonance (SPR) approaches. We show that CRC exists in two major assembly states, low-molecular weight (CRC1; 1[SAT]hexamer + 2[OASS]dimer) and high-molecular weight (CRC2; 1[SAT]hexamer + 4[OASS]dimer) states. Along with AUC results, ITC and SPR studies show that [OASS]dimer binds to [SAT]hexamer in a stepwise manner but the formation of fully saturated CRC3 (1[SAT]hexamer + 6[OASS]dimer) is not favorable. The fraction of CRC2 increases as the [OASS]dimer/[SAT]hexamer ratio increases to >4-fold, but CRC2 can be selectively dissociated into either CRC1 or free enzymes, in the presence of OAS and sulfide, in a concentration-dependent manner. Together, we show that CRC is a regulatable multienzyme assembly, sensitive to OASS-substrate(s) levels but subject to negative cooperativity and steric hindrance. Our results constitute the first report of the dual-assembly-state nature of CRC and suggest that physiological conditions, which limit sulfate uptake, would favor CRC1 over CRC2.


Subject(s)
Cysteine Synthase/chemistry , Cysteine/chemistry , Gene Expression Regulation, Bacterial , Salmonella typhimurium/enzymology , Serine O-Acetyltransferase/chemistry , Binding Sites , Cloning, Molecular , Cysteine/biosynthesis , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salmonella typhimurium/genetics , Serine O-Acetyltransferase/genetics , Serine O-Acetyltransferase/metabolism , Substrate Specificity
12.
Biochemistry ; 56(37): 5011-5025, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28805060

ABSTRACT

By classical competitive antagonism, a substrate and competitive inhibitor must bind mutually exclusively to the active site. The competitive inhibition of O-acetyl serine sulfhydrylase (OASS) by the C-terminus of serine acetyltransferase (SAT) presents a paradox, because the C-terminus of SAT binds to the active site of OASS with an affinity that is 4-6 log-fold (104-106) greater than that of the substrate. Therefore, we employed multiple approaches to understand how the substrate gains access to the OASS active site under physiological conditions. Single-molecule and ensemble approaches showed that the active site-bound high-affinity competitive inhibitor is actively dissociated by the substrate, which is not consistent with classical views of competitive antagonism. We employed fast-flow kinetic approaches to demonstrate that substrate-mediated dissociation of full length SAT-OASS (cysteine regulatory complex) follows a noncanonical "facilitated dissociation" mechanism. To understand the mechanism by which the substrate induces inhibitor dissociation, we resolved the crystal structures of enzyme·inhibitor·substrate ternary complexes. Crystal structures reveal a competitive allosteric binding mechanism in which the substrate intrudes into the inhibitor-bound active site and disengages the inhibitor before occupying the site vacated by the inhibitor. In summary, here we reveal a new type of competitive allosteric binding mechanism by which one of the competitive antagonists facilitates the dissociation of the other. Together, our results indicate that "competitive allostery" is the general feature of noncanonical "facilitated/accelerated dissociation" mechanisms. Further understanding of the mechanistic framework of "competitive allosteric" mechanism may allow us to design a new family of "competitive allosteric drugs/small molecules" that will have improved selectivity and specificity as compared to their competitive and allosteric counterparts.


Subject(s)
Alanine/analogs & derivatives , Bacterial Proteins/antagonists & inhibitors , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/enzymology , Models, Molecular , Salmonella enterica/metabolism , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Alanine/pharmacology , Allosteric Regulation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , Cysteine Synthase/chemistry , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Haemophilus influenzae/metabolism , Kinetics , Ligands , Molecular Conformation , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Oligopeptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Salmonella enterica/enzymology , Serine/chemistry , Serine/metabolism , Serine O-Acetyltransferase/chemistry , Serine O-Acetyltransferase/genetics , Serine O-Acetyltransferase/metabolism , Serine O-Acetyltransferase/pharmacology
13.
Biochim Biophys Acta ; 1854(9): 1184-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25731080

ABSTRACT

O-acetylserine sulfhydrylase A (CysK) is the pyridoxal 5'-phosphate-dependent enzyme that catalyzes the final reaction of cysteine biosynthesis in bacteria. CysK was initially identified in a complex with serine acetyltransferase (CysE), which catalyzes the penultimate reaction in the synthetic pathway. This "cysteine synthase" complex is stabilized by insertion of the CysE C-terminus into the active-site of CysK. Remarkably, the CysK/CysE binding interaction is conserved in most bacterial and plant systems. For the past 40years, CysK was thought to function exclusively in cysteine biosynthesis, but recent studies have revealed a repertoire of additional "moonlighting" activities for this enzyme. CysK and its paralogs influence transcription in both Gram-positive bacteria and the nematode Caenorhabditis elegans. CysK also activates an antibacterial nuclease toxin produced by uropathogenic Escherichia coli. Intriguingly, each moonlighting activity requires a binding partner that invariably mimics the C-terminus of CysE to interact with the CysK active site. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.


Subject(s)
Cysteine Synthase/physiology , Bacteria/metabolism , Binding Sites , Cysteine/biosynthesis , Cysteine Synthase/chemistry , Serine O-Acetyltransferase/chemistry , Serine O-Acetyltransferase/physiology , Transcription, Genetic
14.
Biochim Biophys Acta ; 1854(9): 1175-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25484279

ABSTRACT

The alarming increase of drug resistance in Mycobacterium tuberculosis strains poses a severe threat to human health. Chemotherapy is particularly challenging because M. tuberculosis can persist in the lungs of infected individuals; estimates of the WHO indicate that about 1/3 of the world population is infected with latent tuberculosis providing a large reservoir for relapse and subsequent spread of the disease. Persistent M. tuberculosis shows considerable tolerance towards conventional antibiotics making treatment particularly difficult. In this phase the bacilli are exposed to oxygen and nitrogen radicals generated as part of the host response and redox-defense mechanisms are thus vital for the survival of the pathogen. Sulfur metabolism and de novo cysteine biosynthesis have been shown to be important for the redox homeostasis in persistent M. tuberculosis and these pathways could provide promising targets for novel antibiotics for the treatment of the latent form of the disease. Recent research has provided evidence for three de novo metabolic routes of cysteine biosynthesis in M. tuberculosis, each with a specific PLP dependent cysteine synthase with distinct substrate specificities. In this review we summarize our present understanding of these pathways, with a focus on the advances on functional and mechanistic characterization of mycobacterial PLP dependent cysteine synthases, their role in the various pathways to cysteine, and first attempts to develop specific inhibitors of mycobacterial cysteine biosynthesis. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.


Subject(s)
Cysteine Synthase/chemistry , Mycobacterium tuberculosis/enzymology , Pyridoxal Phosphate/physiology , Anti-Bacterial Agents/pharmacology , Cysteine/biosynthesis , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Humans , Mycobacterium tuberculosis/drug effects
15.
J Enzyme Inhib Med Chem ; 31(sup4): 78-87, 2016.
Article in English | MEDLINE | ID: mdl-27578398

ABSTRACT

Cysteine is a building block for many biomolecules that are crucial for living organisms. O-Acetylserine sulfhydrylase (OASS), present in bacteria and plants but absent in mammals, catalyzes the last step of cysteine biosynthesis. This enzyme has been deeply investigated because, beside the biosynthesis of cysteine, it exerts a series of "moonlighting" activities in bacteria. We have previously reported a series of molecules capable of inhibiting Salmonella typhimurium (S. typhymurium) OASS isoforms at nanomolar concentrations, using a combination of computational and spectroscopic approaches. The cyclopropane-1,2-dicarboxylic acids presented herein provide further insights into the binding mode of small molecules to OASS enzymes. Saturation transfer difference NMR (STD-NMR) was used to characterize the molecule/enzyme interactions for both OASS-A and B. Most of the compounds induce a several fold increase in fluorescence emission of the pyridoxal 5'-phosphate (PLP) coenzyme upon binding to either OASS-A or OASS-B, making these compounds excellent tools for the development of competition-binding experiments.


Subject(s)
Cyclopropanes/pharmacology , Cysteine Synthase/antagonists & inhibitors , Dicarboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Fluorometry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Dicarboxylic Acids/chemical synthesis , Dicarboxylic Acids/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
16.
Biochim Biophys Acta ; 1844(2): 308-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275508

ABSTRACT

O-acetylserine sulfhydrylase (OASS) catalyzes the final step of cysteine biosynthesis from O-acetylserine (OAS) and inorganic sulfide in plants and bacteria. Bioinformatics analyses combined with activity assays enabled us to annotate the two putative genes of Microcystis aeruginosa PCC 7806 to CysK1 and CysK2, which encode the two 75% sequence-identical OASS paralogs. Moreover, we solved the crystal structures of CysK1 at 2.30Ǻ and cystine-complexed CysK2 at 1.91Ǻ, revealing a quite similar overall structure that belongs to the family of fold-type II PLP-dependent enzymes. Structural comparison indicated a significant induced fit upon binding to the cystine, which occupies the binding site for the substrate OAS and blocks the product release tunnel. Subsequent enzymatic assays further confirmed that cystine is a competitive inhibitor of the substrate OAS. Moreover, multiple-sequence alignment revealed that the cystine-binding residues are highly conserved in all OASS proteins, suggesting that this auto-inhibition of cystine might be a universal mechanism of cysteine biosynthesis pathway.


Subject(s)
Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Cysteine/biosynthesis , Feedback, Physiological , Microcystis/enzymology , Amino Acid Sequence , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Cysteine Synthase/genetics , Microcystis/genetics , Models, Molecular , Molecular Sequence Annotation , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
17.
Plant Cell ; 24(6): 2696-706, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22739827

ABSTRACT

Plants produce cyanide (CN-) during ethylene biosynthesis in the mitochondria and require ß-cyanoalanine synthase (CAS) for CN- detoxification. Recent studies show that CAS is a member of the ß-substituted alanine synthase (BSAS) family, which also includes the Cys biosynthesis enzyme O-acetylserine sulfhydrylase (OASS), but how the BSAS evolved distinct metabolic functions is not understood. Here we show that soybean (Glycine max) CAS and OASS form α-aminoacrylate reaction intermediates from Cys and O-acetylserine, respectively. To understand the molecular evolution of CAS and OASS in the BSAS enzyme family, the crystal structures of Gm-CAS and the Gm-CAS K95A mutant with a linked pyridoxal phosphate (PLP)-Cys molecule in the active site were determined. These structures establish a common fold for the plant BSAS family and reveal a substrate-induced conformational change that encloses the active site for catalysis. Comparison of CAS and OASS identified residues that covary in the PLP binding site. The Gm-OASS T81M, S181M, and T185S mutants altered the ratio of OASS:CAS activity but did not convert substrate preference to that of a CAS. Generation of a triple mutant Gm-OASS successfully switched reaction chemistry to that of a CAS. This study provides new molecular insight into the evolution of diverse enzyme functions across the BSAS family in plants.


Subject(s)
Cyanides/pharmacokinetics , Glycine max/metabolism , Lyases/chemistry , Lyases/metabolism , Catalytic Domain , Crystallography, X-Ray , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Inactivation, Metabolic , Lyases/genetics , Models, Molecular , Mutation , Protein Conformation , Glycine max/drug effects , Glycine max/enzymology , Substrate Specificity
18.
Biochim Biophys Acta ; 1830(10): 4573-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23747298

ABSTRACT

BACKGROUND: O-acetyl serine sulfhydrylase (OASS) is a pyridoxal phosphate (PLP) dependent enzyme catalyzing the last step of the cysteine biosynthetic pathway. Here we analyze and investigate the factors responsible for recognition and different conformational changes accompanying the binding of various ligands to OASS. METHODS: X ray crystallography was used to determine the structures of OASS from Entamoeba histolytica in complex with methionine (substrate analog), isoleucine (inhibitor) and an inhibitory tetra-peptide to 2.00Å, 2.03Å and 1.87Å resolutions, respectively. Molecular dynamics simulations were used to investigate the reasons responsible for the extent of domain movement and cleft closure of the enzyme in presence of different ligands. RESULTS: Here we report for the first time an OASS-methionine structure with an unmutated catalytic lysine at the active site. This is also the first OASS structure with a closed active site lacking external aldimine formation. The OASS-isoleucine structure shows the active site cleft in open state. Molecular dynamics studies indicate that cofactor PLP, N88 and G192 form a triad of energy contributors to close the active site upon ligand binding and orientation of the Schiff base forming nitrogen of the ligand is critical for this interaction. CONCLUSIONS: Methionine proves to be a better binder to OASS than isoleucine. The ß branching of isoleucine does not allow it to reorient itself in suitable conformation near PLP to cause active site closure. GENERAL SIGNIFICANCE: Our findings have important implications in designing better inhibitors against OASS across all pathogenic microbial species.


Subject(s)
Cysteine Synthase/metabolism , Entamoeba histolytica/enzymology , Crystallography, X-Ray , Cysteine Synthase/chemistry , Ligands , Models, Molecular , Molecular Dynamics Simulation
19.
Biochim Biophys Acta ; 1834(1): 169-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23000429

ABSTRACT

O-acetylserine sulfhydrylase (OASS) catalyzes the synthesis of l-cysteine in the last step of the reductive sulfate assimilation pathway in microorganisms. Its activity is inhibited by the interaction with serine acetyltransferase (SAT), the preceding enzyme in the metabolic pathway. Inhibition is exerted by the insertion of SAT C-terminal peptide into the OASS active site. This action is effective only on the A isozyme, the prevalent form in enteric bacteria under aerobic conditions, but not on the B-isozyme, the form expressed under anaerobic conditions. We have investigated the active site determinants that modulate the interaction specificity by comparing the binding affinity of thirteen pentapeptides, derived from the C-terminal sequences of SAT of the closely related species Haemophilus influenzae and Salmonella typhimurium, towards the corresponding OASS-A, and towards S. typhimurium OASS-B. We have found that subtle changes in protein active sites have profound effects on protein-peptide recognition. Furthermore, affinity is strongly dependent on the pentapeptide sequence, signaling the relevance of P3-P4-P5 for the strength of binding, and P1-P2 mainly for specificity. The presence of an aromatic residue at P3 results in high affinity peptides with K(diss) in the micromolar and submicromolar range, regardless of the species. An acidic residue, like aspartate at P4, further strengthens the interaction and results in the higher affinity ligand of S. typhimurium OASS-A described to date. Since OASS knocked-out bacteria exhibit a significantly decreased fitness, this investigation provides key information for the development of selective OASS inhibitors, potentially useful as novel antibiotic agents.


Subject(s)
Bacterial Proteins/chemistry , Cysteine Synthase/chemistry , Haemophilus influenzae/enzymology , Salmonella typhimurium/enzymology , Serine O-Acetyltransferase/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Cysteine Synthase/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Peptides/chemistry , Peptides/metabolism , Serine O-Acetyltransferase/metabolism
20.
Biochim Biophys Acta ; 1834(12): 2691-701, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100226

ABSTRACT

O-Acetylserine(thiol)lyases (OAS-TLs) play a pivotal role in a sulfur assimilation pathway incorporating sulfide into amino acids in microorganisms and plants, however, these enzymes have not been found in the animal kingdom. Interestingly, the genome of the roundworm Caenorhabditis elegans contains three expressed genes predicted to encode OAS-TL orthologs (cysl-1-cysl-3), and a related pseudogene (cysl-4); these genes play different roles in resistance to hypoxia, hydrogen sulfide and cyanide. To get an insight into the underlying molecular mechanisms we purified the three recombinant worm OAS-TL proteins, and we determined their enzymatic activities, substrate binding affinities, quaternary structures and the conformations of their active site shapes. We show that the nematode OAS-TL orthologs can bind O-acetylserine and catalyze the canonical reaction although this ligand may more likely serve as a competitive inhibitor to natural substrates instead of being a substrate for sulfur assimilation. In addition, we propose that S-sulfocysteine may be a novel endogenous substrate for these proteins. However, we observed that the three OAS-TL proteins are conformationally different and exhibit distinct substrate specificity. Based on the available evidences we propose the following model: CYSL-1 interacts with EGL-9 and activates HIF-1 that upregulates expression of genes detoxifying sulfide and cyanide, the CYSL-2 acts as a cyanoalanine synthase in the cyanide detoxification pathway and simultaneously produces hydrogen sulfide, while the role of CYSL-3 remains unclear although it exhibits sulfhydrylase activity in vitro. All these data indicate that C. elegans OAS-TL paralogs have distinct cellular functions and may play different roles in maintaining hydrogen sulfide homeostasis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Cysteine Synthase/metabolism , Homeostasis/physiology , Hydrogen Sulfide/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Catalytic Domain , Cell Nucleus/chemistry , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cyanides/metabolism , Cysteine Synthase/chemistry , Cysteine Synthase/genetics , Hydrogen Sulfide/chemistry , Serine/analogs & derivatives , Serine/chemistry , Serine/genetics , Serine/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL