Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Am Chem Soc ; 145(39): 21361-21369, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37733880

ABSTRACT

Apramycin is an aminoglycoside antibiotic isolated from Streptoalloteichus tenebrarius and S. hindustanus that has found clinical use in veterinary medicine. The apramycin structure is notable for its atypical eight-carbon bicyclic dialdose (octose) moiety. While the apramycin biosynthetic gene cluster (apr) has been identified and several of the encoded genes functionally characterized, how the octose core itself is assembled has remained elusive. Nevertheless, recent gene deletion studies have hinted at an N-acetyl aminosugar being a key precursor to the octose, and this hypothesis is consistent with the additional feeding experiments described in the present report. Moreover, bioinformatic analysis indicates that AprG may be structurally similar to GlcNAc-2-epimerase and hence recognize GlcNAc or a structurally similar substrate suggesting a potential role in octose formation. AprG with an extended N-terminal sequence was therefore expressed, purified, and assayed in vitro demonstrating that it does indeed catalyze a transaldolation reaction between GlcNAc or GalNAc and 6'-oxo-lividamine to afford 7'-N-acetyldemethylaprosamine with the same 6'-R and 7'-S stereochemistry as those observed in the apramycin product. Biosynthesis of the octose core in apramycin thus proceeds in the [6 + 2] manner with GlcNAc or GalNAc as the two-carbon donor, which has not been previously reported for biological octose formation, as well as novel inverting stereochemistry of the transferred fragment. Consequently, AprG appears to be a new transaldolase that lacks any apparent sequence similarity to the currently known aldolases and catalyzes a transaldolation for which there is no established biological precedent.


Subject(s)
Nebramycin , Nebramycin/chemistry , Anti-Bacterial Agents , Aminoglycosides , Carbon
2.
ChemMedChem ; 18(1): e202200486, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36198651

ABSTRACT

An intramolecular hydrogen bond between the protonated equatorial 7'-methylamino group of apramycin and the vicinal axial 6'-hydroxy group acidifies the 6'-hydroxy group leading to a strong hydrogen bond to A1408 in the ribosomal drug binding pocket in the decoding A site of the small ribosomal subunit. In 6'-epiapramycin, the trans-nature of the 6'-hydroxy group and the 7'-methylamino group results in a much weaker intramolecular hydrogen bond, and a consequently weaker cooperative hydrogen bonding network with A1408, resulting overall in reduced inhibition of protein synthesis and antibacterial activity.


Subject(s)
Anti-Bacterial Agents , Nebramycin , Hydrogen Bonding , Anti-Bacterial Agents/chemistry , Nebramycin/chemistry , Ribosomes/metabolism , Aminoglycosides
3.
mBio ; 12(1)2021 02 09.
Article in English | MEDLINE | ID: mdl-33563840

ABSTRACT

Apramycin is an aminoglycoside antibiotic with the potential to be developed to combat multidrug-resistant pathogens. Its unique structure evades the clinically widespread mechanisms of aminoglycoside resistance that currently compromise the efficacy of other members in this drug class. Of the aminoglycoside-modifying enzymes that chemically alter these antibiotics, only AAC(3)-IVa has been demonstrated to confer resistance to apramycin through N-acetylation. Knowledge of other modification mechanisms is important to successfully develop apramycin for clinical use. Here, we show that ApmA is structurally unique among the previously described aminoglycoside-modifying enzymes and capable of conferring a high level of resistance to apramycin. In vitro experiments indicated ApmA to be an N-acetyltransferase, but in contrast to AAC(3)-IVa, ApmA has a unique regiospecificity of the acetyl transfer to the N2' position of apramycin. Crystallographic analysis of ApmA conclusively showed that this enzyme is an acetyltransferase from the left-handed ß-helix protein superfamily (LßH) with a conserved active site architecture. The success of apramycin will be dependent on consideration of the impact of this potential form of clinical resistance.IMPORTANCE Apramycin is an aminoglycoside antibiotic that has been traditionally used in veterinary medicine. Recently, it has become an attractive candidate to repurpose in the fight against multidrug-resistant pathogens prioritized by the World Health Organization. Its atypical structure circumvents most of the clinically relevant mechanisms of resistance that impact this class of antibiotics. Prior to repurposing apramycin, it is important to understand the resistance mechanisms that could be a liability. Our study characterizes the most recently identified apramycin resistance element, apmA We show ApmA does not belong to the protein families typically associated with aminoglycoside resistance and is responsible for modifying a different site on the molecule. The data presented will be critical in the development of apramycin derivatives that will evade apmA in the event it becomes prevalent in the clinic.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Escherichia coli/drug effects , Nebramycin/analogs & derivatives , Acetylation , Aminoglycosides/chemistry , Crystallization , Drug Resistance, Bacterial/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Microbial Sensitivity Tests , Nebramycin/chemistry , Nebramycin/metabolism
4.
Angew Chem Int Ed Engl ; 60(16): 8990-8996, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33538390

ABSTRACT

Apramycin is a clinically promising aminoglycoside antibiotic (AGA). To date, mechanisms underlying the biosynthesis and self-resistance of apramycin remain largely unknown. Here we report that apramycin biosynthesis proceeds through unexpected phosphorylation, deacetylation, and dephosphorylation steps, in which a novel aminoglycoside phosphotransferase (AprU), a putative creatinine amidohydrolase (AprP), and an alkaline phosphatase (AprZ) are involved. Biochemical characterization revealed that AprU specifically phosphorylates 5-OH of a pseudotrisaccharide intermediate, whose N-7' acetyl group is subsequently hydrolyzed by AprP. AprZ is located extracellularly where it removes the phosphate group from a pseudotetrasaccharide intermediate, leading to the maturation of apramycin. Intriguingly, 7'-N-acetylated and 5-O-phosphorylated apramycin that were accumulated in ΔaprU and ΔaprZ respectively exhibited significantly reduced antibacterial activities, implying Streptomyces tenebrarius employs C-5 phosphorylation and N-7' acetylation as two strategies to avoid auto-toxicity. Significantly, this study provides insight into the design of new generation AGAs to circumvent the emergence of drug-resistant pathogens.


Subject(s)
Actinobacteria/metabolism , Anti-Bacterial Agents/biosynthesis , Nebramycin/analogs & derivatives , Actinobacteria/chemistry , Anti-Bacterial Agents/chemistry , Nebramycin/biosynthesis , Nebramycin/chemistry
5.
ChemMedChem ; 16(2): 335-339, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33007139

ABSTRACT

We describe the convergent synthesis of a 5-O-ß-D-ribofuranosyl-based apramycin derivative (apralog) that displays significantly improved antibacterial activity over the parent apramycin against wild-type ESKAPE pathogens. In addition, the new apralog retains excellent antibacterial activity in the presence of the only aminoglycoside modifying enzyme (AAC(3)-IV) acting on the parent, without incurring susceptibility to the APH(3') mechanism that disables other 5-O-ß-D-ribofuranosyl 2-deoxystreptamine type aminoglycosides by phosphorylation at the ribose 5-position. Consistent with this antibacterial activity, the new apralog has excellent 30 nM activity (IC50 ) for the inhibition of protein synthesis by the bacterial ribosome in a cell-free translation assay, while retaining the excellent across-the-board selectivity of the parent for inhibition of bacterial over eukaryotic ribosomes. Overall, these characteristics translate into excellent in vivo efficacy against E. coli in a mouse thigh infection model and reduced ototoxicity vis à vis the parent in mouse cochlear explants.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cochlea/drug effects , Escherichia coli/drug effects , Nebramycin/analogs & derivatives , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carbohydrate Conformation , Cochlea/metabolism , Mice , Microbial Sensitivity Tests , Nebramycin/chemical synthesis , Nebramycin/chemistry , Nebramycin/pharmacology
6.
J Am Chem Soc ; 142(1): 530-544, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31790244

ABSTRACT

Apramycin is a structurally unique member of the 2-deoxystreptamine class of aminoglycoside antibiotics characterized by a monosubstituted 2-deoxystreptamine ring that carries an unusual bicyclic eight-carbon dialdose moiety. Because of its unusual structure, apramycin is not susceptible to the most prevalent mechanisms of aminoglycoside resistance including the aminoglycoside-modifying enzymes and the ribosomal methyltransferases whose widespread presence severely compromises all aminoglycosides in current clinical practice. These attributes coupled with minimal ototoxocity in animal models combine to make apramycin an excellent starting point for the development of next-generation aminoglycoside antibiotics for the treatment of multidrug-resistant bacterial infections, particularly the ESKAPE pathogens. With this in mind, we describe the design, synthesis, and evaluation of three series of apramycin derivatives, all functionalized at the 5-position, with the goals of increasing the antibacterial potency without sacrificing selectivity between bacterial and eukaryotic ribosomes and of overcoming the rare aminoglycoside acetyltransferase (3)-IV class of aminoglycoside-modifying enzymes that constitutes the only documented mechanism of antimicrobial resistance to apramycin. We show that several apramycin-5-O-ß-d-ribofuranosides, 5-O-ß-d-eryrthofuranosides, and even simple 5-O-aminoalkyl ethers are effective in this respect through the use of cell-free translation assays with wild-type bacterial and humanized bacterial ribosomes and of extensive antibacterial assays with wild-type and resistant Gram negative bacteria carrying either single or multiple resistance determinants. Ex vivo studies with mouse cochlear explants confirm the low levels of ototoxicity predicted on the basis of selectivity at the target level, while the mouse thigh infection model was used to demonstrate the superiority of an apramycin-5-O-glycoside in reducing the bacterial burden in vivo.


Subject(s)
Aminoacyltransferases/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Glycosides/chemistry , Nebramycin/analogs & derivatives , Anti-Bacterial Agents/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Ethers/chemistry , Microbial Sensitivity Tests , Nebramycin/chemistry , Nebramycin/pharmacology
7.
J Am Chem Soc ; 139(41): 14611-14619, 2017 10 18.
Article in English | MEDLINE | ID: mdl-28892368

ABSTRACT

The preparation of a series of four analogues of the aminoglycoside antibiotics neomycin and paromomycin is described in which ring I, involved in critical binding interactions with the ribosomal target, is replaced by an apramycin-like dioxabicyclo[4.4.0]octane system. The effect of this modification is to lock the hydroxymethyl side chain of the neomycin or paromomycin ring I, as part of the dioxabicyclooctane ring, into either the gauche-gauche or the gauche-trans conformation (respectively, axial or equatorial to the bicyclic system). The antiribosomal activity of these compounds is investigated with cell-free translation assays using both bacterial ribosomes and recombinant hybrid ribosomes carrying eukaryotic decoding A site cassettes. Compounds substituted with an equatorial hydroxyl or amino group in the newly formed ring are considerably more active than their axial diastereomers, lending strong support to crystallographically derived models of aminoglycoside-ribosome interactions. One such bicyclic compound carrying an equatorial hydroxyl group has activity equal to that of the parent yet displays better ribosomal selectivity, predictive of an enhanced therapeutic index. A paromomycin analog lacking the hydroxymethyl ring I side chain is considerably less active than the parent. Antibacterial activity against model Gram negative and Gram positive bacteria is reported for selected compounds, as is activity against ESKAPE pathogens and recombinant bacteria carrying specific resistance determinants. Analogues with a bicyclic ring I carrying equatorial amino or hydroxyl groups mimicking the bound side chains of neomycin and paromomycin, respectively, show excellent activity and, by virtue of their novel structure, retain this activity in strains that are insensitive to the parent compounds.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Nebramycin/analogs & derivatives , Paromomycin/analogs & derivatives , Nebramycin/chemical synthesis , Nebramycin/chemistry , Neomycin/analogs & derivatives , Paromomycin/chemical synthesis , Ribosomes/metabolism
8.
J Am Chem Soc ; 138(20): 6427-35, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27120352

ABSTRACT

Apramycin is a clinically interesting aminoglycoside antibiotic (AGA) containing a highly unique bicyclic octose moiety, and this octose is deoxygenated at the C3 position. Although the biosynthetic pathways for most 2-deoxystreptamine-containing AGAs have been well characterized, the pathway for apramycin biosynthesis, including the C3 deoxygenation process, has long remained unknown. Here we report detailed investigation of apramycin biosynthesis by a series of genetic, biochemical and bioinformatical studies. We show that AprD4 is a novel radical S-adenosyl-l-methionine (SAM) enzyme, which uses a noncanonical CX3CX3C motif for binding of a [4Fe-4S] cluster and catalyzes the dehydration of paromamine, a pseudodisaccharide intermediate in apramycin biosynthesis. We also show that AprD3 is an NADPH-dependent reductase that catalyzes the reduction of the dehydrated product from AprD4-catalyzed reaction to generate lividamine, a C3' deoxygenated product of paromamine. AprD4 and AprD3 do not form a tight catalytic complex, as shown by protein complex immunoprecipitation and other assays. The AprD4/AprD3 enzyme system acts on different pseudodisaccharide substrates but does not catalyze the deoxygenation of oxyapramycin, an apramycin analogue containing a C3 hydroxyl group on the octose moiety, suggesting that oxyapramycin and apramycin are partitioned into two parallel pathways at an early biosynthetic stage. Functional dissection of the C6 dehydrogenase AprQ shows the crosstalk between different AGA biosynthetic gene clusters from the apramycin producer Streptomyces tenebrarius, and reveals the remarkable catalytic versatility of AprQ. Our study highlights the intriguing chemistry in apramycin biosynthesis and nature's ingenuity in combinatorial biosynthesis of natural products.


Subject(s)
Nebramycin/analogs & derivatives , Oxygen/chemistry , Aminoglycosides/metabolism , Carbohydrate Sequence , Catalysis , Nebramycin/biosynthesis , Nebramycin/chemistry , Nebramycin/metabolism , Oxidoreductases/metabolism , Substrate Specificity
9.
Chemistry ; 22(15): 5350-62, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-26928593

ABSTRACT

MicroRNAs (miRNAs) are a recently discovered category of small RNA molecules that regulate gene expression at the post-transcriptional level. Accumulating evidence indicates that miRNAs are aberrantly expressed in a variety of human cancers and that the inhibition of these oncogenic miRNAs could find application in the therapy of different types of cancer. Herein, we describe the synthesis and biological evaluation of new small-molecule drugs that target oncogenic miRNAs production. In particular, we chose to target two miRNAs (i.e., miRNA-372 and -373) implicated in various types of cancer, such as gastric cancer. Their precursors (pre-miRNAs) are overexpressed in cancer cells and lead to mature miRNAs after cleavage of their stem-loop structure by the enzyme Dicer in the cytoplasm. Some of the newly synthesized conjugates can inhibit Dicer processing of the targeted pre-miRNAs in vitro with increased efficacy relative to our previous results (D.D. Vo et al., ACS Chem. Biol. 2014, 9, 711-721) and, more importantly, to inhibit proliferations of adenocarcinoma gastric cancer (AGS) cells overexpressing these miRNAs, thus representing promising leads for future drug development.


Subject(s)
Aminoglycosides/chemistry , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Stomach Neoplasms/chemistry , Biological Evolution , Drug Delivery Systems , Humans , MicroRNAs/chemistry , Models, Molecular , Nebramycin/analogs & derivatives , Nebramycin/chemistry , Neomycin/chemistry , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Structure-Activity Relationship
10.
Talanta ; 146: 423-9, 2016.
Article in English | MEDLINE | ID: mdl-26695285

ABSTRACT

A sensitive non-derivatization method was developed for the analysis of apramycin and impurites using hydrophilic interaction liquid chromatography (HILIC) and charged aerosol detection (CAD). Sample was pretreated with an effective SPE method (recovery >90%) to remove interference with apramycin impurities from sulfate, then analyzed with direct injection. Different chromatography modes of separation and choices of HILIC column were investigated in search of a direct analysis method. The HILIC-CAD method was optimized using a cysteine-bonded zwitterionic HILIC column and compared to the strong cation exchange-ultraviolet (SCX-UV) method with post-column derivatization recommended by the Chinese Pharmacopoeia (veterinary) 2010. The improved chromatographic resolution and peak shape with the HILIC-charged aerosol detection method allows for increase of sample load to 48.9 µg from only 2.8 µg with the SCX-UV approach. More than 16 impurities were detected with this method with improved resolution, and four were identified with MS, while only 7 impurities were detected with the SCX-UV method. Moreover, the current method has a good precision and reproducibility. The intra-day and inter-day of peak area variability was less than or equal to 4.760% RSD and 9.950%, respectively. The average limit of detection and quantization was 80 ng and 200 ng injected on the column, respectively. The overall results demonstrated that the presented method can be used as an alternative to SCX-UV method in the analysis of apramycin and impurities.


Subject(s)
Chromatography, Liquid/methods , Hydrophobic and Hydrophilic Interactions , Nebramycin/analogs & derivatives , Aerosols , Analytic Sample Preparation Methods , Limit of Detection , Nebramycin/analysis , Nebramycin/chemistry
11.
Biochemistry ; 52(30): 5125-32, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23837529

ABSTRACT

The upsurge in drug-resistant tuberculosis (TB) is an emerging global problem. The increased expression of the enhanced intracellular survival (Eis) protein is responsible for the clinical resistance to aminoglycoside (AG) antibiotics of Mycobacterium tuberculosis . Eis from M. tuberculosis (Eis_Mtb) and M. smegmatis (Eis_Msm) function as acetyltransferases capable of acetylating multiple amines of many AGs; however, these Eis homologues differ in AG substrate preference and in the number of acetylated amine groups per AG. The AG binding cavity of Eis_Mtb is divided into two narrow channels, whereas Eis_Msm contains one large cavity. Five bulky residues lining one of the AG binding channels of Eis_Mtb, His119, Ile268, Trp289, Gln291, and Glu401, have significantly smaller counterparts in Eis_Msm, Thr119, Gly266, Ala287, Ala289, and Gly401, respectively. To identify the residue(s) responsible for AG binding in Eis_Mtb and for the functional differences from Eis_Msm, we have generated single, double, triple, quadruple, and quintuple mutants of these residues in Eis_Mtb by mutating them into their Eis_Msm counterparts, and we tested their acetylation activity with three structurally diverse AGs: kanamycin A (KAN), paromomyin (PAR), and apramycin (APR). We show that penultimate C-terminal residue Glu401 plays a critical role in the overall activity of Eis_Mtb. We also demonstrate that the identities of residues Ile268, Trp289, and Gln291 (in Eis_Mtb nomenclature) dictate the differences between the acetylation efficiencies of Eis_Mtb and Eis_Msm for KAN and PAR. Finally, we show that the mutation of Trp289 in Eis_Mtb into Ala plays a role in APR acetylation.


Subject(s)
Acetyltransferases/metabolism , Aminoglycosides/metabolism , Antibiotics, Antitubercular/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Amino Acid Substitution , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Antibiotics, Antitubercular/chemistry , Antibiotics, Antitubercular/pharmacology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Drug Resistance, Multiple, Bacterial , Kanamycin/chemistry , Kanamycin/metabolism , Kanamycin/pharmacology , Kinetics , Molecular Conformation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Nebramycin/analogs & derivatives , Nebramycin/chemistry , Nebramycin/metabolism , Nebramycin/pharmacology , Paromomycin/chemistry , Paromomycin/metabolism , Paromomycin/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 110(33): 13333-8, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23898171

ABSTRACT

Leishmaniasis, a parasitic disease caused by protozoa of the genus Leishmania, affects millions of people worldwide. Aminoglycosides are mostly known as highly potent, broad-spectrum antibiotics that exert their antibacterial activity by selectively targeting the decoding A site of the bacterial ribosome, leading to aberrant protein synthesis. Recently, some aminoglycosides have been clinically approved and are currently used worldwide for the treatment of leishmaniasis; however the molecular details by which aminoglycosides induce their deleterious effect on Leishmaina is still rather obscure. Based on high conservation of the decoding site among all kingdoms, it is assumed that the putative binding site of these agents in Leishmania is the ribosomal A site. However, although recent X-ray crystal structures of the bacterial ribosome in complex with aminoglycosides shed light on the mechanism of aminoglycosides action as antibiotics, no such data are presently available regarding their binding site in Leishmania. We present crystal structures of two different aminoglycoside molecules bound to a model of the Leishmania ribosomal A site: Geneticin (G418), a potent aminoglycoside for the treatment of leishmaniasis at a 2.65-Å resolution, and Apramycin, shown to be a strong binder to the leishmanial ribosome lacking an antileishmanial activity at 1.4-Å resolution. The structural data, coupled with in vitro inhibition measurements on two strains of Leishmania, provide insight as to the source of the difference in inhibitory activity of different Aminoglycosides. The combined structural and physiological data sets the ground for rational design of new, and more specific, aminoglycoside derivatives as potential therapeutic agents against leishmaniasis.


Subject(s)
Gentamicins/chemistry , Gentamicins/pharmacology , Leishmania/drug effects , Leishmaniasis/drug therapy , Models, Molecular , Ribosomal Proteins/chemistry , Crystallization , Gentamicins/metabolism , Leishmania/growth & development , Molecular Structure , Nebramycin/analogs & derivatives , Nebramycin/chemistry , Nebramycin/metabolism , Nebramycin/pharmacology , Protein Binding , Protein Conformation , Ribosomal Proteins/metabolism
13.
J Inorg Biochem ; 124: 26-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583885

ABSTRACT

Three representatives of the distinct antibiotics groups: amoxicillin, apramycin and ristomycin A were studied regarding their impact on hepatitis D virus (HDV) ribozyme both in the metal-free form and complexed with copper(II) ions. Hence the Cu(II)-ristomycin A complex has been characterized by means of NMR, EPR, CD and UV-visible spectroscopic techniques and its binding pattern has been compared with the coordination modes estimated previously for Cu(II)-amoxicillin and Cu(II)-apramycin complexes. It has thus been found that all three antibiotics bind the Cu(II) ion in a very similar manner, engaging two nitrogen and two oxygen donors into coordination with the square planar symmetry in physiological conditions. All three tested antibiotics were able to inhibit the HDV ribozyme catalysis. However, in the presence of the complexes, the catalytic reactions were almost completely inhibited. It was important therefore to check whether the complexes used in lower concentrations could inhibit the HDV ribozyme catalytic activity, thus creating opportunities for their practical application. It turned out that the complexes used in the concentrations of 50µM influenced the catalysis much less effectively comparing to the 200 micromolar concentration. The kobs values were lower than those observed in the control reaction, in the absence of potential inhibitors: 2-fold for amoxicillin, ristomycin A and 3.3-fold for apramycin, respectively.


Subject(s)
Amoxicillin/chemistry , Copper/chemistry , Hepatitis Delta Virus/enzymology , Nebramycin/analogs & derivatives , RNA, Catalytic/chemistry , RNA, Viral/chemistry , Ristocetin/chemistry , Catalysis , Electron Spin Resonance Spectroscopy , Magnetic Resonance Spectroscopy , Nebramycin/chemistry
14.
Proc Natl Acad Sci U S A ; 109(27): 10984-9, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22699498

ABSTRACT

Aminoglycosides are potent antibacterials, but therapy is compromised by substantial toxicity causing, in particular, irreversible hearing loss. Aminoglycoside ototoxicity occurs both in a sporadic dose-dependent and in a genetically predisposed fashion. We recently have developed a mechanistic concept that postulates a key role for the mitochondrial ribosome (mitoribosome) in aminoglycoside ototoxicity. We now report on the surprising finding that apramycin, a structurally unique aminoglycoside licensed for veterinary use, shows little activity toward eukaryotic ribosomes, including hybrid ribosomes which were genetically engineered to carry the mitoribosomal aminoglycoside-susceptibility A1555G allele. In ex vivo cultures of cochlear explants and in the in vivo guinea pig model of chronic ototoxicity, apramycin causes only little hair cell damage and hearing loss but it is a potent antibacterial with good activity against a range of clinical pathogens, including multidrug-resistant Mycobacterium tuberculosis. These data provide proof of concept that antibacterial activity can be dissected from aminoglycoside ototoxicity. Together with 3D structures of apramycin-ribosome complexes at 3.5-Å resolution, our results provide a conceptual framework for further development of less toxic aminoglycosides by hypothesis-driven chemical synthesis.


Subject(s)
Aminoglycosides/toxicity , Bacterial Infections/drug therapy , Deafness/chemically induced , Mitochondria/drug effects , Nebramycin/analogs & derivatives , Ribosomes/drug effects , Animals , Anti-Bacterial Agents/toxicity , Bacteria/drug effects , Binding Sites/drug effects , Deafness/physiopathology , Drug Design , Gentamicins/toxicity , Guinea Pigs , HEK293 Cells , Hair Cells, Auditory/drug effects , Humans , Mice , Mitochondria/metabolism , Mutagenesis/physiology , Mycobacterium/drug effects , Nebramycin/chemistry , Nebramycin/toxicity , Organ Culture Techniques , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Pseudomonas aeruginosa/drug effects , Rabbits , Reticulocytes/cytology , Ribosomes/chemistry , Ribosomes/metabolism , Staphylococcus aureus/drug effects
15.
Anal Chim Acta ; 661(1): 76-84, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20113718

ABSTRACT

The development of an efficient analytical protocol for the reliable identification of the biosynthetic intermediates found in microbial cultures, which usually produce complex intermediates of the metabolites of interest, is both challenging and essential for further studies into gene-to-metabolite networks. A simple and highly selective method for detecting the biosynthetic intermediates involved in the aminoglycosidic nebramycin pathway of Streptomyces tenebrarius was developed and validated. Cleanup utilizing a solid-phase extraction (OASIS MCX SPE) technique provides a simple and reproducible method for extracting the nebramycin factors from a fermentation broth. The separation of each factor through a reversed-phase C(18) column using an ion-pairing reagent allowed the simultaneous profiling of the aminoglycosides. By employing the authentic tobramycin spiked into a blank fermentation medium, the combined use of acid extraction, OASIS MCX SPE cleanup, and HFBA (heptafluorobutyric acid)-mediated chromatographic separation coupled with electrospray ionization-tandem mass spectrometry (ESI-MS/MS) detection was proven to be sufficiently accurate and reliable to analyze the nebramycin factors produced in a culture broth. The detection limit of tobramycin spiked in the culture broth was approximately 1.8 ng mL(-1). The mean recovery ranged from 89 to 92%, the intra- and inter-day precision (RSD) was <6% and their accuracy ranged from 88 to 93%. A total of nine nebramycin factors including apramycin, 6''-O-carbamoylkanamycin B, 6''-O-carbamoyltobramycin, 3'-hydoxyapramycin, tobramycin, kanamycin B, NK-1012-1, nebramine, and neamine were identified. This is the first report on the integrated LC-ESI-MS/MS characterization of a wide range of nebramycin factors from a bacterial fermentation broth.


Subject(s)
Nebramycin/chemistry , Streptomyces/chemistry , Tobramycin/chemistry , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Chromatography, Liquid , Molecular Structure , Spectrometry, Mass, Electrospray Ionization , Streptomyces/classification
17.
Dalton Trans ; (7): 1123-30, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19322482

ABSTRACT

The interaction of apramycin with copper at different pH values was investigated by potentiometric titrations and EPR, UV-vis and CD spectroscopic techniques. The Cu(II)-apramycin complex prevailing at pH 6.5 was further characterized by NMR spectroscopy. Metal-proton distances derived from paramagnetic relaxation enhancements were used as restraints in a conformational search procedure in order to define the structure of the complex. Longitudinal relaxation rates were measured with the IR-COSY pulse sequence, thus solving the problems due to signal overlap. At pH 6.5 apramycin binds copper(II) with a 2 : 1 stoichiometry, through the vicinal hydroxyl and deprotonated amino groups of ring III. Plasmid DNA electrophoresis showed that the Cu(II)-apramycin complex is more active than free Cu(II) in generating strand breakages. Interestingly, this complex in the presence of ascorbic acid damages DNA with a higher yield than in the presence of H(2)O(2).


Subject(s)
Copper/chemistry , DNA/chemistry , Nebramycin/analogs & derivatives , Oxidative Stress/drug effects , Plasmids/chemistry , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Circular Dichroism , Copper/metabolism , DNA/metabolism , Electron Spin Resonance Spectroscopy , Hydrogen Peroxide/pharmacology , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Chemical , Nebramycin/chemistry , Nebramycin/metabolism , Oxidants/pharmacology , Thermodynamics
18.
Nucleic Acids Res ; 35(21): 7128-39, 2007.
Article in English | MEDLINE | ID: mdl-17942426

ABSTRACT

Owing to a striking, and most likely fortuitous, structural and sequence similarity with the bacterial 16 S ribosomal A site, the RNA kissing-loop complex formed by the HIV-1 genomic RNA dimerization initiation site (DIS) specifically binds 4,5-disubstituted 2-deoxystreptamine (2-DOS) aminoglycoside antibiotics. We used chemical probing, molecular modeling, isothermal titration calorimetry (ITC) and UV melting to investigate aminoglycoside binding to the DIS loop-loop complex. We showed that apramycin, an aminoglycoside containing a bicyclic moiety, also binds the DIS, but in a different way than 4,5-disubstituted 2-DOS aminoglycosides. The determination of thermodynamic parameters for various aminoglycosides revealed the role of the different rings in the drug-RNA interaction. Surprisingly, we found that the affinity of lividomycin and neomycin for the DIS (K(d) approximately 30 nM) is significantly higher than that obtained in the same experimental conditions for their natural target, the bacterial A site (K(d) approximately 1.6 microM). In good agreement with their respective affinity, aminoglycoside increase the melting temperature of the loop-loop interaction and also block the conversion from kissing-loop complex to extended duplex. Taken together, our data might be useful for selecting new molecules with improved specificity and affinity toward the HIV-1 DIS RNA.


Subject(s)
5' Untranslated Regions/chemistry , Aminoglycosides/chemistry , Antiviral Agents/chemistry , HIV-1/genetics , RNA, Viral/chemistry , Anti-Bacterial Agents/chemistry , Binding Sites , Calorimetry , Cinnamates/chemistry , Dimerization , Hygromycin B/analogs & derivatives , Hygromycin B/chemistry , Models, Molecular , Nebramycin/analogs & derivatives , Nebramycin/chemistry , Nucleic Acid Conformation , Paromomycin/analogs & derivatives , Paromomycin/chemistry , Thermodynamics
19.
J AOAC Int ; 90(4): 885-91, 2007.
Article in English | MEDLINE | ID: mdl-17760324

ABSTRACT

A high-performance liquid chromatographic method for determining apramycin in animal feeds was developed. Apramycin in feeds was extracted with 0.1 M HCl solution and cleaned up with an MCX solid-phase extraction column. The purified extract was derivatized with o-phthaldehyde, and components were separated on a C18 column and detected with a fluorescence detector. Mass spectrometric data confirmed that apramycin was derivatized at all the 4 primary amines on the apramycin molecule. Average recoveries at 8 included levels (5, 10, 20, 40, 80, 200, 400, and 2000 mg/kg) ranged from 92.2 to 100.5%, and the coefficients of variation were < 6.5%. Standard curves were linear over the range 0.05 to 10 microg/mL. The detection and quantitation limits were determined to be 0.2 and 1.0 mg/kg, respectively.


Subject(s)
Chromatography, Liquid/methods , Nebramycin/analogs & derivatives , Spectrometry, Fluorescence/methods , Animal Feed , Buffers , Chemistry, Pharmaceutical/methods , Mass Spectrometry/methods , Models, Chemical , Nebramycin/analysis , Nebramycin/chemistry , Reproducibility of Results , Silanes/chemistry , Solid Phase Extraction , Time Factors
20.
Chembiochem ; 8(14): 1700-9, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17705310

ABSTRACT

The lack of absolute prokaryotic selectivity of natural antibiotics is widespread and is a significant clinical problem. The use of this disadvantage of aminoglycoside antibiotics for the possible treatment of human genetic diseases is extremely challenging. Here, we have used a combination of biochemical and structural analysis to compare and contrast the molecular mechanisms of action and the structure-activity relationships of a new synthetic aminoglycoside, NB33, and a structurally similar natural aminoglycoside apramycin. The data presented herein demonstrate the general molecular principles that determine the decreased selectivity of apramycin for the prokaryotic decoding site, and the increased selectivity of NB33 for the eukaryotic decoding site. These results are therefore extremely beneficial for further research on both the design of new aminoglycoside-based antibiotics with diminished deleterious effects on humans, as well as the design of new aminoglycoside-based structures that selectively target the eukaryotic ribosome.


Subject(s)
Aminoglycosides/chemistry , Nebramycin/analogs & derivatives , Paromomycin/analogs & derivatives , Base Sequence , Binding Sites , Crystallography, X-Ray/methods , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Kinetics , Luciferases/metabolism , Models, Chemical , Molecular Conformation , Molecular Sequence Data , Nebramycin/chemistry , Nucleic Acid Conformation , Paromomycin/chemistry , Paromomycin/pharmacology , Protein Binding , RNA/chemistry , Ribosomes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...