Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Structure ; 25(5): 750-761.e5, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28416110

ABSTRACT

The macrolides are a class of antibiotic, characterized by a large macrocyclic lactone ring that can be inactivated by macrolide phosphotransferase enzymes. We present structures for MPH(2')-I and MPH(2')-II in the apo state, and in complex with GTP analogs and six different macrolides. These represent the first structures from the two main classes of macrolide phosphotransferases. The structures show that the enzymes are related to the aminoglycoside phosphotransferases, but are distinguished from them by the presence of a large interdomain linker that contributes to an expanded antibiotic binding pocket. This pocket is largely hydrophobic, with a negatively charged patch located at a conserved aspartate residue, rationalizing the broad-spectrum resistance conferred by the enzymes. Complementary mutation studies provide insights into factors governing substrate specificity. A comparison with macrolides bound to their natural target, the 50S ribosome, suggests avenues for next-generation antibiotic development.


Subject(s)
Bacterial Proteins/chemistry , Drug Resistance, Bacterial , Macrolides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Bacteria/drug effects , Bacteria/enzymology , Bacterial Proteins/metabolism , Binding Sites , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Macrolides/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Substrate Specificity
2.
Article in English | MEDLINE | ID: mdl-23805415

ABSTRACT

Aminoglycosides are a class of antibiotics with a broad spectrum of antimicrobial activity. Unfortunately, resistance in clinical isolates is pervasive, rendering many aminoglycosides ineffective. The most widely disseminated means of resistance to this class of antibiotics is inactivation of the drug by aminoglycoside-modifying enzymes (AMEs). There are two principal strategies to overcoming the effects of AMEs. The first approach involves the design of novel aminoglycosides that can evade modification. Although this strategy has yielded a number of superior aminoglycoside variants, their efficacy cannot be sustained in the long term. The second approach entails the development of molecules that interfere with the mechanism of AMEs such that the activity of aminoglycosides is preserved. Although such a molecule has yet to enter clinical development, the search for AME inhibitors has been greatly facilitated by the wealth of structural information amassed in recent years. In particular, aminoglycoside phosphotransferases or kinases (APHs) have been studied extensively and crystal structures of a number of APHs with diverse regiospecificity and substrate specificity have been elucidated. In this review, we present a comprehensive overview of the available APH structures and recent progress in APH inhibitor development, with a focus on the structure-guided strategies.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enzyme Inhibitors/isolation & purification , Kanamycin Kinase/chemistry , Kanamycin Kinase/metabolism , Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/enzymology , Drug Discovery/methods , Enzyme Inhibitors/metabolism , Kanamycin Kinase/antagonists & inhibitors , Models, Molecular , Protein Conformation
3.
PLoS One ; 6(5): e19589, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21573013

ABSTRACT

Antibiotic resistance is recognized as a growing healthcare problem. To address this issue, one strategy is to thwart the causal mechanism using an adjuvant in partner with the antibiotic. Aminoglycosides are a class of clinically important antibiotics used for the treatment of serious infections. Their usefulness has been compromised predominantly due to drug inactivation by aminoglycoside-modifying enzymes, such as aminoglycoside phosphotransferases or kinases. These kinases are structurally homologous to eukaryotic Ser/Thr and Tyr protein kinases and it has been shown that some can be inhibited by select protein kinase inhibitors. The aminoglycoside kinase, APH(3')-IIIa, can be inhibited by CKI-7, an ATP-competitive inhibitor for the casein kinase 1. We have determined that CKI-7 is also a moderate inhibitor for the atypical APH(9)-Ia. Here we present the crystal structures of CKI-7-bound APH(3')-IIIa and APH(9)-Ia, the first structures of a eukaryotic protein kinase inhibitor in complex with bacterial kinases. CKI-7 binds to the nucleotide-binding pocket of the enzymes and its binding alters the conformation of the nucleotide-binding loop, the segment homologous to the glycine-rich loop in eukaryotic protein kinases. Comparison of these structures with the CKI-7-bound casein kinase 1 reveals features in the binding pockets that are distinct in the bacterial kinases and could be exploited for the design of a bacterial kinase specific inhibitor. Our results provide evidence that an inhibitor for a subset of APHs can be developed in order to curtail resistance to aminoglycosides.


Subject(s)
Eukaryotic Cells/metabolism , Isoquinolines/chemistry , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Binding Sites , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/chemistry , Casein Kinase I/metabolism , Crystallography, X-Ray , Drug Design , Kanamycin Kinase/antagonists & inhibitors , Kanamycin Kinase/chemistry , Kanamycin Kinase/metabolism , Nucleotides/chemistry , Nucleotides/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding/drug effects
4.
J Biol Chem ; 285(13): 9545-9555, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20089863

ABSTRACT

Aminoglycoside phosphotransferases (APHs) constitute a diverse group of enzymes that are often the underlying cause of aminoglycoside resistance in the clinical setting. Several APHs have been extensively characterized, including the elucidation of the three-dimensional structure of two APH(3') isozymes and an APH(2'') enzyme. Although many APHs are plasmid-encoded and are capable of inactivating numerous 2-deoxystreptmaine aminoglycosides with multiple regiospecificity, APH(9)-Ia, isolated from Legionella pneumophila, is an unusual enzyme among the APH family for its chromosomal origin and its specificity for a single non-2-deoxystreptamine aminoglycoside substrate, spectinomycin. We describe here the crystal structures of APH(9)-Ia in its apo form, its binary complex with the nucleotide, AMP, and its ternary complex bound with ADP and spectinomycin. The structures reveal that APH(9)-Ia adopts the bilobal protein kinase-fold, analogous to the APH(3') and APH(2'') enzymes. However, APH(9)-Ia differs significantly from the other two types of APH enzymes in its substrate binding area and that it undergoes a conformation change upon ligand binding. Moreover, kinetic assay experiments indicate that APH(9)-Ia has stringent substrate specificity as it is unable to phosphorylate substrates of choline kinase or methylthioribose kinase despite high structural resemblance. The crystal structures of APH(9)-Ia demonstrate and expand our understanding of the diversity of the APH family, which in turn will facilitate the development of new antibiotics and inhibitors.


Subject(s)
Bacterial Proteins/chemistry , Drug Resistance, Microbial , Legionella pneumophila/enzymology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray/methods , Kinetics , Ligands , Models, Chemical , Models, Molecular , Molecular Conformation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phylogeny , Protein Isoforms , Protein Structure, Tertiary
5.
Antimicrob Agents Chemother ; 53(7): 3049-55, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19433564

ABSTRACT

Butirosin is unique among the naturally occurring aminoglycosides, having a substituted amino group at position 1 (N1) of the 2-deoxystreptamine ring with an (S)-4-amino-2-hydroxybutyrate (AHB) group. While bacterial resistance to aminoglycosides can be ascribed chiefly to drug inactivation by plasmid-encoded aminoglycoside-modifying enzymes, the presence of an AHB group protects the aminoglycoside from binding to many resistance enzymes, and hence, the antibiotic retains its bactericidal properties. Consequently, several semisynthetic N1-substituted aminoglycosides, such as amikacin, isepamicin, and netilmicin, were developed. Unfortunately, butirosin, amikacin, and isepamicin are not resistant to inactivation by 3'-aminoglycoside O-phosphotransferase type IIIa [APH(3')-IIIa]. We report here the crystal structure of APH(3')-IIIa in complex with an ATP analog, AMPPNP [adenosine 5'-(beta,gamma-imido)triphosphate], and butirosin A to 2.4-A resolution. The structure shows that butirosin A binds to the enzyme in a manner analogous to other 4,5-disubstituted aminoglycosides, and the flexible antibiotic-binding loop is key to the accommodation of structurally diverse substrates. Based on the crystal structure, we have also constructed a model of APH(3')-IIIa in complex with amikacin, a commonly used semisynthetic N1-substituted 4,6-disubstituted aminoglycoside. Together, these results suggest a strategy to further derivatize the AHB group in order to generate new aminoglycoside derivatives that can elude inactivation by resistance enzymes while maintaining their ability to bind to the ribosomal A site.


Subject(s)
Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial , Kanamycin Kinase/chemistry , Amikacin/chemistry , Butirosin Sulfate/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Structure, Secondary
6.
Biochim Biophys Acta ; 1764(1): 63-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16344011

ABSTRACT

Integrity of the cell wall is essential for bacterial survival, and as a consequence components involved in its biosynthesis can potentially be exploited as targets for antibiotics. One such potential target is CTP:glycerol-3-phosphate cytidylyltransferase. This enzyme (TarD(Sa) in Staphylococcus aureus and TagD(Bs) in Bacillus subtilis) catalyzes the formation of CDP-glycerol, which is used for the assembly of linkages between peptidoglycan and teichoic acid polymer in Gram-positive bacteria. Intriguingly, despite the high sequence identity between TarD(Sa) and TagD(Bs) (69% identity), kinetic studies show that these two enzymes differ markedly in their kinetic mechanism and activity. To examine the basis for the disparate enzymological properties, we have determined the crystal structure of TarD(Sa) in the apo state to 3 A resolution, and performed equilibrium sedimentation analysis. Comparison of the structure with that of CTP- and CDP-glycerol-bound TagD(Bs) crystal structures reveals that the overall structure of TarD(Sa) is essentially the same as that of TagD(Bs), except in the C-terminus, where it forms a helix in TagD(Bs) but is disordered in the apo TarD(Sa) structure. In addition, TarD(Sa) can exist both as a tetramer and as a dimer, unlike TagD(Bs), which is a dimer. These observations shed light on the structural basis for the differing kinetic characteristics between TarD(Sa) and TagD(Bs).


Subject(s)
Nucleotidyltransferases/chemistry , Staphylococcus aureus/enzymology , Bacillus subtilis/enzymology , Crystallography, X-Ray , Cytidine Triphosphate/metabolism , Kinetics , Models, Molecular , Nucleotidyltransferases/metabolism , Protein Folding , Protein Structure, Quaternary , Substrate Specificity
7.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 10): 1897-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15388945

ABSTRACT

3'-Aminoglycoside kinase type IIIa [APH(3')-IIIa] catalyzes the transfer of gamma-phosphate from ATP to the 3'-hydroxyl of many aminoglycoside antibiotics, abolishing their bactericidal effects. Despite very low sequence identity, APH(3')-IIIa and eukaryotic protein kinases share structural and functional similarities, including a sensitivity to isoquinolinsulfonamide-type inhibitors. APH(3')-IIIa has been cocrystallized with CKI-7, a casein kinase 1 inhibitor. These crystals were grown using PEG 3000 as precipitant and required consecutive cycles of microseeding. Data were collected to 2.5 A. The crystals belong to space group P2(1)2(1)2(1), with unit-cell parameters a = 49.84, b = 91.90, c = 131.2 A.


Subject(s)
Aminoglycosides/chemistry , Crystallography, X-Ray/methods , Isoquinolines/chemistry , Protein Kinases/chemistry , Bacterial Proteins/chemistry , Catalysis , Isoquinolines/pharmacology , Polyethylene Glycols/chemistry , Protein Conformation
8.
EMBO J ; 21(10): 2323-31, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12006485

ABSTRACT

The misuse of antibiotics has selected for bacteria that have evolved mechanisms for evading the effects of these drugs. For aminoglycosides, a group of clinically important bactericidal antibiotics that target the A-site of the 16S ribosomal RNA, the most common mode of resistance is enzyme-catalyzed chemical modification of the drug. While aminoglycosides are structurally diverse, a single enzyme can confer resistance to many of these antibiotics. For example, the aminoglycoside kinase APH(3')-IIIa, produced by pathogenic Gram-positive bacteria such as enterococci and staphylococci, is capable of detoxifying at least 10 distinct aminoglycosides. Here we describe the crystal structures of APH(3')-IIIa in complex with ADP and kanamycin A or neomycin B. These structures reveal that the basis for this enzyme's substrate promiscuity is the presence of two alternative subsites in the antibiotic binding pocket. Furthermore, comparison between the A-site of the bacterial ribosome and APH(3')-IIIa shows that mimicry is the second major factor in dictating the substrate spectrum of APH(3')-IIIa. These results suggest a potential strategy for drug design aimed at circumventing antibiotic resistance.


Subject(s)
Kanamycin Kinase/genetics , Kanamycin Kinase/metabolism , RNA, Ribosomal, 16S/genetics , Crystallography, X-Ray , Gram-Positive Bacteria/enzymology , Gram-Positive Bacteria/genetics , Kanamycin Kinase/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...