Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
ACS Catal ; 10(6): 3548-3555, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-38250052

ABSTRACT

Nucleotidyl transfer is an archetypal enzyme reaction central to DNA replication and repair. Here we describe a variation of the nucleotidylation reaction termed "catch and release" that is used by an antibiotic modifying enzyme. The aminoglycoside nucleotidyl transferase 4' (ANT4') inactivates antibiotics such as kanamycin and neomycin through nucleotidylation within an active site that shares significant structural, and inferred underlying catalytic similarity, with human DNA polymerase beta. Here we follow the entire nucleotidyl transfer reaction coordinate of ANT4' covalently inactivating neomycin using X-ray crystallography. These studies show that although the underlying reaction mechanism is conserved with polymerases, a short 2.35 A hydrogen bond is initially formed to facilitate tight binding of the aminoglycoside substrate and is subsequently disrupted by the assembly of the catalytically active ternary complex. This enables the release of products post catalysis due to a lower free energy of the product state compared to the starting substrate complex. We propose that this "catch and release" mechanism of antibiotic turnover observed in ANT4' is a variation of nucleotidyl transfer that has been adapted for the inactivation of antibiotics.

2.
Angew Chem Int Ed Engl ; 58(45): 16260-16266, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31515870

ABSTRACT

The position, bonding and dynamics of hydrogen atoms in the catalytic centers of proteins are essential for catalysis. The role of short hydrogen bonds in catalysis has remained highly debated and led to establishment of several distinctive geometrical arrangements of hydrogen atoms vis-à-vis the heavier donor and acceptor counterparts, that is, low-barrier, single-well or short canonical hydrogen bonds. Here we demonstrate how the position of a hydrogen atom in the catalytic triad of an aminoglycoside inactivating enzyme leads to a thirty-fold increase in catalytic turnover. A low-barrier hydrogen bond is present in the enzyme active site for the substrates that are turned over the best, whereas a canonical hydrogen bond is found with the least preferred substrate. This is the first comparison of these hydrogen bonds involving an identical catalytic network, while directly demonstrating how active site electrostatics adapt to the electronic nature of substrates to tune catalysis.


Subject(s)
Acetyltransferases/metabolism , Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Acetyltransferases/chemistry , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Binding Sites , Catalysis , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Conformation , Static Electricity
3.
Biochemistry ; 58(12): 1579-1586, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30793594

ABSTRACT

The aminoglycoside nucleotidyltransferase(4') is an enzyme with high substrate promiscuity and catalyzes the transfer of the AMP group from ATP to the 4'-OH site of many structurally diverse aminoglycosides, which results in the elimination of their effectiveness as antibiotics. Two thermostable variants carrying single-site mutations are used to determine the molecular properties associated with thermophilicity. The thermodynamics of enzyme-ligand interactions showed that one variant (T130K) has properties identical to those of the mesophilic wild type (WT) while the other (D80Y) behaved differently. Differences between D80Y and the T130K/WT pair include the change in heat capacity (Δ Cp), which is dependent on temperature for D80Y but not for WT or T130K. The change in Δ Cp with temperature (ΔΔ Cp) with D80Y is dependent on aminoglycoside only in H2O and remains the same with all aminoglycosides in D2O. Furthermore, the offset temperature ( Toff), the temperature difference that yields identical enthalpies in H2O and D2O, becomes larger with an increase in temperature for WT and T130K but remains mostly unchanged for D80Y. Studies in H2O and D2O revealed that solvent reorganization becomes the major contributor to ligand binding with an increase in temperature for WT and T130K, but changes in low-frequency vibrational modes are the main contributors with D80Y. Data presented in this paper suggest that global properties associated with the enzyme-ligand interactions, such as the thermodynamics of ligand binding, may yield clues about thermophilicity and permit us to distinguish those variants that are simply a more thermostable version of the mesophilic protein.


Subject(s)
Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Nucleotidyltransferases/metabolism , Cyanobacteria/enzymology , Escherichia coli/genetics , Geobacillus stearothermophilus/enzymology , Ligands , Protein Binding , Protein Isoforms/metabolism , Staphylococcus aureus/enzymology , Temperature , Thermodynamics , Thermosynechococcus
4.
J Med Chem ; 61(22): 10218-10227, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30347146

ABSTRACT

Aminoglycoside antibiotics are a large family of antibiotics that can be divided into two distinct classes on the basis of the substitution pattern of the central deoxystreptamine ring. Although aminoglycosides are chemically, structurally, and topologically diverse, some aminoglycoside-modifying enzymes (AGMEs) are able to inactivate as many as 15 aminoglycosides from the two main classes, the kanamycin- and neomycin-based antibiotics. Here, we present the crystal structure of a promiscuous AGME, aminoglycoside- N3-acetyltransferase-IIIb (AAC-IIIb), in the apo form, in binary drug (sisomicin, neomycin, and paromomycin) and coenzyme A (CoASH) complexes, and in the ternary neomycin-CoASH complex. These data provide a structural framework for interpretation of the thermodynamics of enzyme-ligand interactions and the role of solvent in the recognition of ligands. In combination with the recent structure of an AGME that does not have broad substrate specificity, these structures allow for the direct determination of how antibiotic promiscuity is encoded in some AGMEs.


Subject(s)
Acetyltransferases/metabolism , Acetyltransferases/chemistry , Amino Acid Sequence , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , Binding Sites , Ligands , Models, Molecular , Protein Conformation , Solvents/chemistry , Substrate Specificity , Thermodynamics
5.
Sci Adv ; 4(4): eaas8667, 2018 04.
Article in English | MEDLINE | ID: mdl-29632894

ABSTRACT

One group of enzymes that confer resistance to aminoglycoside antibiotics through covalent modification belongs to the GCN5-related N-acetyltransferase (GNAT) superfamily. We show how a unique GNAT subfamily member uses a previously unidentified noncanonical catalytic triad, consisting of a glutamic acid, a histidine, and the antibiotic substrate itself, which acts as a nucleophile and attacks the acetyl donor molecule. Neutron diffraction studies allow for unambiguous identification of a low-barrier hydrogen bond, predicted in canonical catalytic triads to increase basicity of the histidine. This work highlights the role of this unique catalytic triad in mediating antibiotic resistance while providing new insights into the design of the next generation of aminoglycosides.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial , Hydrogen Bonding , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Catalysis , Crystallography, X-Ray , Drug Design , Models, Molecular , Molecular Conformation , Neutrons , Structure-Activity Relationship , Temperature
6.
J Phys Chem B ; 121(29): 7086-7094, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28689415

ABSTRACT

Aminoglycoside nucleotidyltransferase 4' (ANT) is a homodimeric enzyme that modifies the C4'-OH site of aminoglycoside antibiotics by nucleotidylation. A few single- and double-residue mutants of this enzyme (T130K, D80Y, and D80Y/T130K) from Bacillus stearothermophilus show increased thermostability. This article investigates how such residue replacements, which are distant from the active site and monomer-monomer interface, result in various changes of the thermostability of the enzyme. In this work, we show that the thermodynamic properties of enzyme-ligand complexes and protein dynamics may be indicators of a thermophilic behavior. Our data suggests that one of the single-site mutants of ANT, D80Y, may be a thermophilic protein and the other thermostable mutant, T130K, is actually a more heat-stable variant of the mesophilic wild type (WT) with a higher Tm. Our data also suggest that T130K and D80Y adopt different global dynamics strategies to achieve different levels of thermostability enhancement and that the differences between the properties of the species can be described in terms of global dynamics rather than in terms of specific structural features. Thermophilicity of the D80Y comes at the cost of less favorable thermodynamic parameters for ligand binding relative to WT. On the other hand, the T130K species exhibits the same affinity to ligands and the same thermodynamic parameters of complex formation as the WT enzyme. These observations suggest that a quantitative characterization of ligand binding and protein dynamics can be used to differentiate thermophilic proteins from their simply more heat-stable mesophilic counterparts.


Subject(s)
Geobacillus stearothermophilus/enzymology , Temperature , Enzyme Stability/genetics , Geobacillus stearothermophilus/genetics , Hot Temperature , Ligands , Mutation , Thermodynamics
7.
Proteins ; 85(7): 1258-1265, 2017 07.
Article in English | MEDLINE | ID: mdl-28316100

ABSTRACT

Kinetic, thermodynamic, and structural properties of the aminoglycoside N3-acetyltransferase-VIa (AAC-VIa) are determined. Among the aminoglycoside N3-acetyltransferases, AAC-VIa has one of the most limited substrate profiles. Kinetic studies showed that only five aminoglycosides are substrates for this enzyme with a range of fourfold difference in kcat values. Larger differences in KM (∼40-fold) resulted in ∼30-fold variation in kcat /KM . Binding of aminoglycosides to AAC-VIa was enthalpically favored and entropically disfavored with a net result of favorable Gibbs energy (ΔG < 0). A net deprotonation of the enzyme, ligand, or both accompanied the formation of binary and ternary complexes. This is opposite of what was observed with several other aminoglycoside N3-acetyltransferases, where ligand binding causes more protonation. The change in heat capacity (ΔCp) was different in H2 O and D2 O for the binary enzyme-sisomicin complex but remained the same in both solvents for the ternary enzyme-CoASH-sisomicin complex. Unlike, most other aminoglycoside-modifying enzymes, the values of ΔCp were within the expected range of protein-carbohydrate interactions. Solution behavior of AAC-VIa was also different from the more promiscuous aminoglycoside N3-acetyltransferases and showed a monomer-dimer equilibrium as detected by analytical ultracentrifugation (AUC). Binding of ligands shifted the enzyme to monomeric state. Data also showed that polar interactions were the most dominant factor in dimer formation. Overall, thermodynamics of ligand-protein interactions and differences in protein behavior in solution provide few clues on the limited substrate profile of this enzyme despite its >55% sequence similarity to the highly promiscuous aminoglycoside N3-acetyltransferase. Proteins 2017; 85:1258-1265. © 2017 Wiley Periodicals, Inc.


Subject(s)
Acetyltransferases/chemistry , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Enterobacter cloacae/chemistry , Protons , Sisomicin/chemistry , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amino Acid Motifs , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Deuterium Oxide/chemistry , Enterobacter cloacae/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gentamicins/chemistry , Gentamicins/metabolism , Kanamycin/chemistry , Kanamycin/metabolism , Kinetics , Ligands , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sisomicin/metabolism , Structural Homology, Protein , Substrate Specificity , Thermodynamics , Tobramycin/chemistry , Tobramycin/metabolism , Water/chemistry
8.
Biochim Biophys Acta ; 1860(4): 802-13, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26802312

ABSTRACT

BACKGROUND: Aminoglycoside O-phosphotransferases make up a large class of bacterial enzymes that is widely distributed among pathogens and confer a high resistance to several clinically used aminoglycoside antibiotics. Aminoglycoside 2″-phosphotransferase IVa, APH(2″)-IVa, is an important member of this class, but there is little information on the thermodynamics of aminoglycoside binding and on the nature of its rate-limiting step. METHODS: We used isothermal titration calorimetry, electrostatic potential calculations, molecular dynamics simulations and X-ray crystallography to study the interactions between the enzyme and different aminoglycosides. We determined the rate-limiting step of the reaction by the means of transient kinetic measurements. RESULTS: For the first time, Kd values were determined directly for APH(2″)-IVa and different aminoglycosides. The affinity of the enzyme seems to anti-correlate with the molecular weight of the ligand, suggesting a limited degree of freedom in the binding site. The main interactions are electrostatic bonds between the positively charged amino groups of aminoglycosides and Glu or Asp residues of APH. In spite of the significantly different ratio Kd/Km, there is no large difference in the transient kinetics obtained with the different aminoglycosides. We show that a product release step is rate-limiting for the overall reaction. CONCLUSIONS: APH(2″)-IVa has a higher affinity for aminoglycosides carrying an amino group in 2' and 6', but tighter bindings do not correlate with higher catalytic efficiencies. As with APH(3')-IIIa, an intermediate containing product is preponderant during the steady state. GENERAL SIGNIFICANCE: This intermediate may constitute a good target for future drug design.


Subject(s)
Aminoglycosides/chemistry , Bacterial Proteins/chemistry , Enterococcus/enzymology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Aminoglycosides/metabolism , Bacterial Proteins/metabolism , Kinetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Static Electricity
9.
Biochemistry ; 54(32): 5120, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26252172

ABSTRACT

Biochemistry 2012, 51 (45), 9147−9155. DOI: 10.1021/bi301126g. Page 9148. A corrected version of the Figure 2 legend appears here: Figure 2. Backbone of the ANT D80Y variant in ribbon representation. Two monomer subunits are colored red and green. Bound kanamycin A molecules are colored blue, and Mg-AMPCPP molecules are colored yellow (Protein Data Bank entry 1KNY).14 Page 9148 (last line). The sentence should read, "A thermostable variant of ANT, T130K, was obtained from thermophilic cyanobacterium T. elongatus."


Subject(s)
Aminoglycosides/metabolism , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Cyanobacteria/enzymology , Cyanobacteria/genetics , Drug Resistance, Microbial , Enzyme Stability , Genetic Variation , Nucleotidyltransferases/genetics , Thermodynamics
10.
Biochemistry ; 53(34): 5544-50, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25093604

ABSTRACT

The aminoglycoside nucleotidyltransferase-4' (ANT) is an enzyme that causes resistance to a large number of aminoglycoside antibiotics by nucleotidylation of the 4'-site on these antibiotics. The effect of solvent reorganization on enzyme-ligand interactions was investigated using a thermophilic variant of the enzyme resulting from a single-site mutation (T130K). Data showed that the binding of aminoglycosides to ANT causes exposure of polar groups to solvent. However, solvent reorganization becomes the major contributor to the enthalpy of the formation of enzyme-aminoglycoside complexes only above 20 °C. The change in heat capacity (ΔCp) shows an aminoglycoside-dependent pattern such that it correlates with the affinity of the ligand for the enzyme. Differences in ΔCp values determined in H2O and D2O also correlated with the ligand affinity. The temperature-dependent increase in the offset temperature (Toff), the temperature difference required to observe equal enthalpies in both solvents, is also dependent on the binding affinity of the ligand, and the steepest increase was observed with the tightest binding aminoglycoside, neomycin. Overall, these data, together with earlier observations with a different enzyme, the aminoglycoside N3-acetyltransferase-IIIb [Norris, A. L., and Serpersu, E. H. (2011) Biochemistry 50, 9309], show that solvent reorganization or changes in soft vibrational modes of the protein are interchangeable with respect to the role of being the major contributor to complex formation depending on temperature. These data suggest that such effects may more generally apply to enzyme-ligand interactions, and studies at a single temperature may provide only a part of the whole picture of thermodynamics of enzyme-ligand interactions.


Subject(s)
Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Nucleotidyltransferases/chemistry , Solvents/chemistry , Calorimetry , Enzyme Stability , Hot Temperature , Thermodynamics
11.
Biochemistry ; 53(1): 30-8, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24320996

ABSTRACT

The aminoglycoside N3 acetyltransferase-IIIb (AAC) is responsible for conferring bacterial resistance to a variety of aminoglycoside antibiotics. Nuclear magnetic resonance spectroscopy and dynamic light scattering analyses revealed a surprising result; the dynamics of the ternary complex between AAC and its two ligands, an antibiotic and coenzyme A, are dependent upon the order in which the ligands are bound. Additionally, two structurally similar aminoglycosides, neomycin and paromomycin, induce strikingly different dynamic properties when they are in their ternary complexes. To the best of our knowledge, this is the first example of a system in which two identically productive pathways of forming a simple ternary complex yield significant differences in dynamic properties. These observations emphasize the importance of the sequence of events in achieving optimal protein-ligand interactions and demonstrate that even a minor difference in molecular structure can have a profound effect on biochemical processes.


Subject(s)
Acetyltransferases/chemistry , Coenzyme A/chemistry , Neomycin/chemistry , Paromomycin/chemistry , Aminoglycosides/chemistry , Ligands , Light , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Scattering, Radiation , Thermodynamics
12.
Protein Sci ; 22(7): 916-28, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640799

ABSTRACT

Aminoglycoside-modifying enzymes (AGMEs) are expressed in many pathogenic bacteria and cause resistance to aminoglycoside (AG) antibiotics. Remarkably, the substrate promiscuity of AGMEs is quite variable. The molecular basis for such ligand promiscuity is largely unknown as there is not an obvious link between amino acid sequence or structure and the antibiotic profiles of AGMEs. To address this issue, this article presents the first kinetic and thermodynamic characterization of one of the least promiscuous AGMEs, the AG N3 acetyltransferase-IIa (AAC-IIa) and its comparison to two highly promiscuous AGMEs, the AG N3-acetyltransferase-IIIb (AAC-IIIb) and the AG phosphotransferase(3')-IIIa (APH). Despite having similar antibiotic selectivities, AAC-IIIb and APH catalyze different reactions and share no homology to one another. AAC-IIa and AAC-IIIb catalyze the same reaction and are very similar in both amino acid sequence and structure. However, they demonstrate strong differences in their substrate profiles and kinetic and thermodynamic properties. AAC-IIa and APH are also polar opposites in terms of ligand promiscuity but share no sequence or apparent structural homology. However, they both are highly dynamic and may even contain disordered segments and both adopt well-defined conformations when AGs are bound. Contrary to this AAC-IIIb maintains a well-defined structure even in apo form. Data presented herein suggest that the antibiotic promiscuity of AGMEs may be determined neither by the flexibility of the protein nor the size of the active site cavity alone but strongly modulated or controlled by the effects of the cosubstrate on the dynamic and thermodynamic properties of the enzyme.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Acetylation , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Calorimetry , Kinetics , Ligands , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Substrate Specificity , Thermodynamics
13.
FEBS Lett ; 586(23): 4223-7, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23108046

ABSTRACT

Aminoglycoside phosphotransferases are bacterial enzymes responsible for the inactivation of aminoglycoside antibiotics by O-phosphorylation. It is important to understand the mechanism of enzymes in order to find efficient drugs. Using rapid-mixing methods, we studied the transient kinetics of aminoglycoside phosphotransferase(3')-IIIa. We show that an ADP-enzyme complex is the main steady state intermediate. This intermediate interacts strongly with kanamycin A to form an abortive complex that traps the enzyme in an inactive state. A good strategy to prevent the inactivation of aminoglycosides would be to develop uncompetitive inhibitors that interact with this key ADP-enzyme complex.


Subject(s)
Kanamycin Kinase/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial , Kanamycin/metabolism , Kanamycin/pharmacology , Kinetics
14.
Biochemistry ; 51(45): 9147-55, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23066871

ABSTRACT

The aminoglycoside nucleotidyltransferase (4') (ANT) is an aminoglycoside-modifying enzyme that detoxifies antibiotics by nucleotidylating at the C4'-OH site. Previous crystallographic studies show that the enzyme is a homodimer and each subunit binds one kanamycin and one Mg-AMPCPP, where the transfer of the nucleotidyl group occurs between the substrates bound to different subunits. In this work, sedimentation velocity analysis of ANT by analytical ultracentrifugation showed the enzyme exists as a mixture of a monomer and a dimer in solution and that dimer formation is driven by hydrophobic interactions between the subunits. The binding of aminoglycosides shifts the equilibrium toward dimer formation, while the binding of the cosubstrate, Mg-ATP, has no effect on the monomer-dimer equilibrium. Surprisingly, binding of several divalent cations, including Mg(2+), Mn(2+), and Ca(2+), to the enzyme also shifted the equilibrium in favor of dimer formation. Binding studies, performed by electron paramagnetic resonance spectroscopy, showed that divalent cations bind to the aminoglycoside binding site in the absence of substrates with a stoichiometry of 2:1. Energetic aspects of binding of all aminoglycosides to ANT were determined by isothermal titration calorimetry to be enthalpically favored and entropically disfavored with an overall favorable Gibbs energy. Aminoglycosides in the neomycin class each bind to the enzyme with significantly different enthalpic and entropic contributions, while those of the kanamycin class bind with similar thermodynamic parameters.


Subject(s)
Nucleotidyltransferases/chemistry , Aminoglycosides/metabolism , Binding Sites , Calorimetry , Cations, Divalent/metabolism , Drug Resistance, Microbial , Kanamycin/metabolism , Neomycin/metabolism , Nucleotidyltransferases/metabolism , Protein Multimerization , Ribostamycin/metabolism , Thermodynamics
16.
Biochemistry ; 51(11): 2309-18, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22369433

ABSTRACT

Previous osmotic stress studies on the role of solvent in two structurally unrelated dihydrofolate reductases (DHFRs) found weaker binding of dihydrofolate (DHF) to either enzyme in the presence of osmolytes. To explain these unusual results, weak interactions between DHF and osmolytes were proposed, with a competition between osmolyte and DHFR for DHF. High osmolyte concentrations will inhibit binding of the cognate pair. To evaluate this hypothesis, we devised a small molecule approach. Dimerization of folate, monitored by nuclear magnetic resonance, was weakened 2-3-fold upon addition of betaine or dimethyl sulfoxide (DMSO), supporting preferential interaction of either osmolyte with the monomer (as it possesses a larger surface area). Nuclear Overhauser effect (NOE) spectroscopy experiments found a positive NOE for the interaction of the C3'/C5' benzoyl ring protons with the C9 proton in buffer; however, a negative NOE was observed upon addition of betaine or DMSO. This change indicated a decreased tumbling rate, consistent with osmolyte interaction. Osmotic stress experiments also showed that betaine, DMSO, and sucrose preferentially interact with folate. Further, studies with the folate fragments, p-aminobenzoic acid and pterin 6-carboxylate, revealed interactions for both model compounds with betaine and sucrose. In contrast, DMSO was strongly excluded from the pterin ring but preferentially interacted with the p-aminobenzoyl moiety. These interactions are likely to be important in vivo because of the crowded conditions of the cell where weak contacts can more readily compete with specific binding interactions.


Subject(s)
Folic Acid/analogs & derivatives , 4-Aminobenzoic Acid/chemistry , 4-Aminobenzoic Acid/metabolism , Binding Sites , Dimerization , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Kinetics , Osmolar Concentration , Pteridines , Solutions , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism
17.
Biochemistry ; 50(48): 10559-65, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22026726

ABSTRACT

NMR spectroscopy experiments and molecular dynamics simulations were performed to describe the dynamic properties of the aminoglycoside acetyltransferase (3)-IIIb (AAC) in its apo and coenzyme A (CoASH) bound forms. The (15)N-(1)H HSQC spectra indicate a partial structural change and coupling of the CoASH binding site with another region in the protein upon the CoASH titration into the apo enzyme. Molecular dynamics simulations indicate a significant structural and dynamic variation of the long loop in the antibiotic binding domain in the form of a relatively slow (250 ns), concerted opening motion in the CoASH-enzyme complex and that binding of the CoASH increases the structural flexibility of the loop, leading to an interchange between several similar equally populated conformations.


Subject(s)
Acetyltransferases/chemistry , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Bacillus anthracis/enzymology , Coenzyme A/chemistry , Molecular Dynamics Simulation , Apoproteins/chemistry , Binding Sites , Magnetic Resonance Spectroscopy , Protein Binding , Protein Conformation , Static Electricity , Substrate Specificity , Thermodynamics
18.
Biochemistry ; 50(43): 9309-17, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21958034

ABSTRACT

The results presented here show the first known observation of opposite signs of change in heat capacity (ΔC(p)) of two structurally similar ligands binding to the same protein site. Neomycin and paromomycin are aminoglycoside antibiotics that are substrates for the resistance-conferring enzyme, the aminoglycoside acetyltransferase-(3)-IIIb (AAC). These antibiotics are identical to one another except at the 6' position where neomycin has an amine and paromomycin has a hydroxyl. The opposite trends in ΔC(p) of binding of these two drugs to AAC suggest a differential exposure of nonpolar amino acid side chains. Nuclear magnetic resonance experiments further demonstrate significantly different changes in AAC upon interaction with neomycin and paromomycin. Experiments in H(2)O and D(2)O reveal the first observed temperature dependence of solvent and vibrational contributions to ΔC(p). Coenzyme A significantly influences these effects. Together, the data suggest that AAC exploits solvent properties to facilitate favorable thermodynamic selection of antibiotics.


Subject(s)
Acetyltransferases/metabolism , Anti-Bacterial Agents/metabolism , Escherichia coli/enzymology , Neomycin/metabolism , Paromomycin/metabolism , Binding Sites , Calorimetry , Coenzyme A/metabolism , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Solvents/metabolism , Substrate Specificity , Thermodynamics
19.
J Mol Biol ; 409(3): 450-65, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21477597

ABSTRACT

The bacterial enzyme aminoglycoside phosphotransferase(3')-IIIa (APH) confers resistance against a wide range of aminoglycoside antibiotics. In this study, we use the Gaussian network model to investigate how the binding of nucleotides and antibiotics influences the dynamics and thereby the ligand binding properties of APH. Interestingly, in NMR experiments, the dynamics differ significantly in various APH complexes, although crystallographic studies indicate that no larger conformational changes occur upon ligand binding. Isothermal titration calorimetry also shows different thermodynamic contributions to ligand binding. Formation of aminoglycoside-APH complexes is enthalpically driven, while the enthalpic change upon aminoglycoside binding to the nucleotide-APH complex is much smaller. The differential effects of nucleotide binding and antibiotic binding to APH can be explained theoretically by single-residue fluctuations and correlated motions of the enzyme. The surprising destabilization of ß-sheet residues upon nucleotide binding, as seen in hydrogen/deuterium exchange experiments, shows that the number of closest neighbors does not fully explain residue flexibility. Additionally, we must consider correlated motions of dynamic protein domains, which show that not only connectivity but also the overall protein architecture is important for protein dynamics.


Subject(s)
Adenosine Triphosphate/chemistry , Anti-Bacterial Agents/chemistry , Drug Resistance, Bacterial , Kanamycin Kinase/chemistry , Metabolic Networks and Pathways , Protein Binding , Substrate Specificity , Thermodynamics
20.
Biochemistry ; 49(19): 4027-35, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20387903

ABSTRACT

The thermodynamic and kinetic properties of interactions of antibiotics with the aminoglycoside acetyltransferase (3)-IIIb (AAC) are determined with several experimental methods. These data represent the first such characterization of an enzyme that modifies the 2-deoxystreptamine ring common to all aminoglycoside antibiotics. Antibiotic substrates for AAC include kanamycin A, kanamycin B, tobramycin, sisomicin, neomycin B, paromomycin, lividomycin A, and ribostamycin. Kinetic studies show that kanamycin group aminoglycosides have higher k(cat) values than members of the neomycin group. Only small aminoglycosides without intraring constraints show substrate inhibition. Isothermal titration calorimetry (ITC) and fluorescence measurements are consistent with a molecular size-dependent stoichiometry where binding stoichiometries are 1.5-2.0 for small antibiotics and 1.0 for larger. Antibiotic-enzyme interaction occurs with a favorable enthalpy (DeltaH < 0) and a compensating unfavorable entropy (TDeltaS < 0). The presence of coenzyme A significantly increases the affinity of the antibiotic for AAC. However, the thermodynamic properties of its ternary complexes distinguish this enzyme from other aminoglycoside-modifying enzymes (AGMEs). Unlike other AGMEs, the enthalpy of binding becomes more favored by 1.7-10.0-fold in the presence of the cosubstrate CoASH, while the entropy becomes 2.0-22.5-fold less favored. The overall free energy change is still only 1.0-1.9 kcal/mol from binary to ternary for all antibiotics tested, which is similar to those for other aminoglycoside-modifying enzymes. A computationally derived homology model provides structural support for these conclusions and further indicates that AAC is likely a member of the GCN5-related acetyltransferase family of proteins.


Subject(s)
Acetyltransferases/chemistry , Anti-Bacterial Agents/chemistry , Acetyltransferases/antagonists & inhibitors , Binding Sites , Kanamycin/analogs & derivatives , Kanamycin/chemistry , Kinetics , Paromomycin/chemistry , Ribostamycin/chemistry , Substrate Specificity , Thermodynamics , Tobramycin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...