Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 874
Filter
1.
Proc Natl Acad Sci U S A ; 120(12): e2217254120, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36917671

ABSTRACT

The potentiation of antibiotics is a promising strategy for combatting antibiotic-resistant/tolerant bacteria. Herein, we report that a 5-min sublethal heat shock enhances the bactericidal actions of aminoglycoside antibiotics by six orders of magnitude against both exponential- and stationary-phase Escherichia coli. This combined treatment also effectively kills various E. coli persisters, E. coli clinical isolates, and numerous gram-negative but not gram-positive bacteria and enables aminoglycosides at 5% of minimum inhibitory concentrations to eradicate multidrug-resistant pathogens Acinetobacter baumannii and Klebsiella pneumoniae. Mechanistically, the potentiation is achieved comprehensively by heat shock-enhanced proton motive force that thus promotes the bacterial uptake of aminoglycosides, as well as by increasing irreversible protein aggregation and reactive oxygen species that further augment the downstream lethality of aminoglycosides. Consistently, protonophores, chemical chaperones, antioxidants, and anaerobic culturing abolish heat shock-enhanced aminoglycoside lethality. We also demonstrate as a proof of concept that infrared irradiation- or photothermal nanosphere-induced thermal treatments potentiate aminoglycoside killing of Pseudomonas aeruginosa in a mouse acute skin wound model. Our study advances the understanding of the mechanism of actions of aminoglycosides and demonstrates a high potential for thermal ablation in curing bacterial infections when combined with aminoglycosides.


Subject(s)
Aminoglycosides , Anti-Bacterial Agents , Mice , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Reactive Oxygen Species/pharmacology , Protein Aggregates , Escherichia coli , Gram-Negative Bacteria , Bacteria , Heat-Shock Response , Microbial Sensitivity Tests
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35064089

ABSTRACT

Kasugamycin (KSG) is an aminoglycoside antibiotic widely used in agriculture and exhibits considerable medical potential. Previous studies suggested that KSG interferes with translation by blocking binding of canonical messenger RNA (mRNA) and initiator transfer tRNA (tRNA) to the small ribosomal subunit, thereby preventing initiation of protein synthesis. Here, by using genome-wide approaches, we show that KSG can interfere with translation even after the formation of the 70S initiation complex on mRNA, as the extent of KSG-mediated translation inhibition correlates with increased occupancy of start codons by 70S ribosomes. Even at saturating concentrations, KSG does not completely abolish translation, allowing for continuing expression of some Escherichia coli proteins. Differential action of KSG significantly depends on the nature of the mRNA residue immediately preceding the start codon, with guanine in this position being the most conducive to inhibition by the drug. In addition, the activity of KSG is attenuated by translational coupling as genes whose start codons overlap with the coding regions or the stop codons of the upstream cistrons tend to be less susceptible to drug-mediated inhibition. Altogether, our findings reveal KSG as an example of a small ribosomal subunit-targeting antibiotic with a well-pronounced context specificity of action.


Subject(s)
Aminoglycosides/pharmacology , Binding Sites , Peptide Chain Initiation, Translational/drug effects , RNA, Messenger/genetics , Ribosomes/metabolism , Aminoglycosides/chemistry , Codon, Initiator , Molecular Structure , Open Reading Frames , Protein Binding , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribosomes/chemistry , Structure-Activity Relationship
3.
BMC Genomics ; 24(1): 120, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36927548

ABSTRACT

BACKGROUND: The aminoglycosides are established antibiotics that inhibit bacterial protein synthesis by binding to ribosomal RNA. Additional non-antibiotic aminoglycoside cellular functions have also been identified through aminoglycoside interactions with cellular RNAs. The full extent, however, of genome-wide aminoglycoside RNA interactions in Escherichia coli has not been determined. Here, we report genome-wide identification and verification of the aminoglycoside Kanamycin B binding to Escherichia coli RNAs. Immobilized Kanamycin B beads in pull-down assays were used for transcriptome-profiling analysis (RNA-seq). RESULTS: Over two hundred Kanamycin B binding RNAs were identified. Functional classification analysis of the RNA sequence related genes revealed a wide range of cellular functions. Small RNA fragments (ncRNA, tRNA and rRNA) or small mRNA was used to verify the binding with Kanamycin B in vitro. Kanamycin B and ibsC mRNA was analysed by chemical probing. CONCLUSIONS: The results will provide biochemical evidence and understanding of potential extra-antibiotic cellular functions of aminoglycosides in Escherichia coli.


Subject(s)
Escherichia coli , RNA , RNA/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , RNA, Ribosomal/chemistry , RNA, Messenger/genetics
4.
RNA ; 27(1): 12-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33028652

ABSTRACT

Identifying small molecules that selectively bind an RNA target while discriminating against all other cellular RNAs is an important challenge in RNA-targeted drug discovery. Much effort has been directed toward identifying drug-like small molecules that minimize electrostatic and stacking interactions that lead to nonspecific binding of aminoglycosides and intercalators to many stem-loop RNAs. Many such compounds have been reported to bind RNAs and inhibit their cellular activities. However, target engagement and cellular selectivity assays are not routinely performed, and it is often unclear whether functional activity directly results from specific binding to the target RNA. Here, we examined the propensities of three drug-like compounds, previously shown to bind and inhibit the cellular activities of distinct stem-loop RNAs, to bind and inhibit the cellular activities of two unrelated HIV-1 stem-loop RNAs: the transactivation response element (TAR) and the rev response element stem IIB (RREIIB). All compounds bound TAR and RREIIB in vitro, and two inhibited TAR-dependent transactivation and RRE-dependent viral export in cell-based assays while also exhibiting off-target interactions consistent with nonspecific activity. A survey of X-ray and NMR structures of RNA-small molecule complexes revealed that aminoglycosides and drug-like molecules form hydrogen bonds with functional groups commonly accessible in canonical stem-loop RNA motifs, in contrast to ligands that specifically bind riboswitches. Our results demonstrate that drug-like molecules can nonspecifically bind stem-loop RNAs most likely through hydrogen bonding and electrostatic interactions and reinforce the importance of assaying for off-target interactions and RNA selectivity in vitro and in cells when assessing novel RNA-binders.


Subject(s)
Aminoglycosides/pharmacology , Genes, env/drug effects , HIV Long Terminal Repeat/drug effects , RNA, Viral/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Base Pairing , Base Sequence , Binding Sites , Biological Assay , Drug Discovery , HIV-1/drug effects , HIV-1/genetics , HIV-1/metabolism , Humans , Hydrogen Bonding , Isoquinolines/chemistry , Isoquinolines/metabolism , Isoquinolines/pharmacology , Nucleic Acid Conformation , Pentamidine/chemistry , Pentamidine/metabolism , Pentamidine/pharmacology , RNA, Viral/genetics , RNA, Viral/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Static Electricity , Transcriptional Activation/drug effects , Yohimbine/chemistry , Yohimbine/metabolism , Yohimbine/pharmacology
5.
Cell ; 135(4): 679-90, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19013277

ABSTRACT

Aminoglycoside antibiotics, such as gentamicin and kanamycin, directly target the ribosome, yet the mechanisms by which these bactericidal drugs induce cell death are not fully understood. Recently, oxidative stress has been implicated as one of the mechanisms whereby bactericidal antibiotics kill bacteria. Here, we use systems-level approaches and phenotypic analyses to provide insight into the pathway whereby aminoglycosides ultimately trigger hydroxyl radical formation. We show, by disabling systems that facilitate membrane protein traffic, that mistranslation and misfolding of membrane proteins are central to aminoglycoside-induced oxidative stress and cell death. Signaling through the envelope stress-response two-component system is found to be a key player in this process, and the redox-responsive two-component system is shown to have an associated role. Additionally, we show that these two-component systems play a general role in bactericidal antibiotic-mediated oxidative stress and cell death, expanding our understanding of the common mechanism of killing induced by bactericidal antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/metabolism , Membrane Proteins/drug effects , Protein Biosynthesis/drug effects , Aminoglycosides/chemistry , Escherichia coli/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Hydroxyl Radical , Models, Biological , Models, Genetic , Oxidation-Reduction , Oxidative Stress , Protein Denaturation , Protein Folding
6.
Mol Cell ; 58(5): 832-44, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26028538

ABSTRACT

The increase in multi-drug-resistant bacteria is limiting the effectiveness of currently approved antibiotics, leading to a renewed interest in antibiotics with distinct chemical scaffolds. We have solved the structures of the Thermus thermophilus 70S ribosome with A-, P-, and E-site tRNAs bound and in complex with either the aminocyclitol-containing antibiotic hygromycin A (HygA) or the nucleoside antibiotic A201A. Both antibiotics bind at the peptidyl transferase center and sterically occlude the CCA-end of the A-tRNA from entering the A site of the peptidyl transferase center. Single-molecule Förster resonance energy transfer (smFRET) experiments reveal that HygA and A201A specifically interfere with full accommodation of the A-tRNA, leading to the presence of tRNA accommodation intermediates and thereby inhibiting peptide bond formation. Thus, our results provide not only insight into the mechanism of action of HygA and A201A, but also into the fundamental process of tRNA accommodation during protein synthesis.


Subject(s)
Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Cinnamates/chemistry , Hygromycin B/analogs & derivatives , RNA, Transfer/chemistry , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Cinnamates/pharmacology , Crystallography, X-Ray , Disk Diffusion Antimicrobial Tests , Drug Resistance, Bacterial , Escherichia coli/drug effects , Hydrogen Bonding , Hygromycin B/chemistry , Hygromycin B/pharmacology , Models, Molecular , Protein Conformation , Thermus thermophilus
7.
Curr Microbiol ; 81(1): 5, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37950074

ABSTRACT

LanK is a TetR type regulatory protein that coordinates the late steps of the biosynthesis of the landomycin family of antitumor angucyclic polyketides and their export from the cells of Streptomyces cyanogenus S136. We recently described the structure of LanK and showed that it is the carbohydrate portion of the landomycins that is responsible for abrogating the repressing effect of LanK on landomycin production and export. The effect has been established in a series of in vitro tests using synthetic analogs of the landomycin carbohydrate chains. Whether such synthetic compounds would function as effector molecules for LanK under in vivo conditions remained unknown. Furthermore, the location and identity of LanK operator sites within the lanK-lanJ intergenic region (lanKJp) was unknown. Here we report that methoxyphenyl analogs of tri- and hexasaccharide chains of landomycins cannot function as LanK ligands when applied externally to the reporter strain. The lability of these compounds to cellular media and/or their poor penetration into the cells could explain our observations. The LanK operator site has been mapped to a 14-bp region of lanKJp that includes a plausible -35 site upstream of the lanK start codon in a series of electrophoretic DNA mobility shift assays. This opens the door to studies of the DNA-LanK interaction at a single nucleotide resolution level.


Subject(s)
Aminoglycosides , Transcription Factors , Aminoglycosides/chemistry , Transcription Factors/genetics , DNA
8.
Proc Natl Acad Sci U S A ; 117(48): 30423-30432, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199626

ABSTRACT

Rifampicin (Rif) is a first-line therapeutic used to treat the infectious disease tuberculosis (TB), which is caused by the pathogen Mycobacterium tuberculosis (Mtb). The emergence of Rif-resistant (RifR) Mtb presents a need for new antibiotics. Rif targets the enzyme RNA polymerase (RNAP). Sorangicin A (Sor) is an unrelated inhibitor that binds in the Rif-binding pocket of RNAP. Sor inhibits a subset of RifR RNAPs, including the most prevalent clinical RifR RNAP substitution found in Mtb infected patients (S456>L of the ß subunit). Here, we present structural and biochemical data demonstrating that Sor inhibits the wild-type Mtb RNAP by a similar mechanism as Rif: by preventing the translocation of very short RNAs. By contrast, Sor inhibits the RifR S456L enzyme at an earlier step, preventing the transition of a partially unwound promoter DNA intermediate to the fully opened DNA and blocking the template-strand DNA from reaching the active site in the RNAP catalytic center. By defining template-strand blocking as a mechanism for inhibition, we provide a mechanistic drug target in RNAP. Our finding that Sor inhibits the wild-type and mutant RNAPs through different mechanisms prompts future considerations for designing antibiotics against resistant targets. Also, we show that Sor has a better pharmacokinetic profile than Rif, making it a suitable starting molecule to design drugs to be used for the treatment of TB patients with comorbidities who require multiple medications.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antitubercular/pharmacology , DNA-Directed RNA Polymerases/metabolism , Drug Resistance, Bacterial/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/physiology , Promoter Regions, Genetic , Aminoglycosides/chemistry , Antibiotics, Antitubercular/chemistry , Binding Sites , Humans , Models, Molecular , Molecular Conformation , Protein Binding , Rifampin/pharmacology , Structure-Activity Relationship , Tuberculosis/drug therapy , Tuberculosis/microbiology
9.
Angew Chem Int Ed Engl ; 62(3): e202213810, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36411245

ABSTRACT

Herein, bioinspired total syntheses of A201A, A201D, and A201E based on a previously reported biosynthetic pathway are presented. The challenging 1,2-cis-furanoside, a core structure of the A201 family, was obtained by remote 2-quinolinecarbonyl-assisted glycosylation. We accomplished the total synthesis of A201A and A201E based on the critical 1,2-cis-furanoside moiety through late-stage glycosylation without any interference from basic dimethyl adenosine. We also confirmed the absolute configuration of A201E by total synthesis. This modular synthesis strategy enables efficient preparation of A201 family antibiotics, allowing the study of their structure-activity relationships and mode of action. This study satisfies the increasing demand for developing novel antibiotics inspired by the A201 family.


Subject(s)
Anti-Bacterial Agents , Nucleosides , Aminoglycosides/chemistry , Glycosylation
10.
Antimicrob Agents Chemother ; 66(12): e0099222, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36346250

ABSTRACT

Pseudomonas aeruginosa is an opportunistic Gram-negative bacterium that causes nosocomial infections in immunocompromised patients. ß-lactam and aminoglycoside antibiotics are commonly used in the treatment of P. aeruginosa infections. Previously, we found that mutation in a PA4292 gene increases bacterial resistance to ß-lactam antibiotics. In this study, we demonstrated that mutation in PA4292 increases bacterial susceptibility to aminoglycoside antibiotics. We further found enhanced uptake of tobramycin by the ΔPA4292 mutant, which might be due to an increase of proton motive force (PMF). Sequence analysis revealed PA4292 is homologous to the Escherichia coli phosphate transporter PitA. Mutation of PA4292 indeed reduces intracellular phosphate concentration. We thus named PA4292 as pitA. Although the PMF is enhanced in the ΔpitA mutant, the intracellular ATP concentration is lower than that in the isogenic wild-type strain PA14, which might be due to lack of the ATP synthesis substrate phosphate. Overexpression of the phosphate transporter complex genes pstSCAB in the ΔpitA mutant restores the intracellular phosphate concentration, PMF, ATP synthesis, and aminoglycosides resistance. In addition, growth of wild-type PA14 in a low-phosphate medium resulted in higher PMF and aminoglycoside susceptibility compared to cells grown in a high-phosphate medium. Overall, our results demonstrate the roles of PitA in phosphate transportation and reveal the relationship between intracellular phosphate and aminoglycoside susceptibility.


Subject(s)
Proton-Motive Force , Pseudomonas aeruginosa , Adenosine Triphosphate , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Anti-Bacterial Agents/pharmacology , beta-Lactams , Escherichia coli/genetics , Phosphate Transport Proteins , Phosphates , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism
11.
Antimicrob Agents Chemother ; 66(2): e0112521, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34902270

ABSTRACT

Improving the efficacy of existing antibiotics is a promising strategy for combating antibiotic-resistant/tolerant bacterial pathogens that have become a severe threat to human health. We previously reported that aminoglycoside antibiotics could be dramatically potentiated against stationary-phase Escherichia coli cells under hypoionic shock conditions (i.e., treatment with ion-free solutions), but the underlying molecular mechanism remains unknown. Here, we show that mechanosensitive (MS) channels, a ubiquitous protein family sensing mechanical forces of cell membrane, mediate such hypoionic shock-induced aminoglycoside potentiation. Two-minute treatment under conditions of hypoionic shock (e.g., in pure water) greatly enhances the bactericidal effects of aminoglycosides against both spontaneous and triggered E. coli persisters, numerous strains of Gram-negative pathogens in vitro, and Pseudomonas aeruginosa in mice. Such potentiation is achieved by hypoionic shock-enhanced bacterial uptake of aminoglycosides and is linked to hypoionic shock-induced destabilization of the cytoplasmic membrane in E. coli. Genetic and biochemical analyses reveal that MscS-family channels directly and redundantly mediate aminoglycoside uptake upon hypoionic shock and thus potentiation, with MscL channel showing reduced effect. Molecular docking and site-directed mutagenesis analyses reveal a putative streptomycin-binding pocket in MscS, critical for streptomycin uptake and potentiation. These results suggest that hypoionic shock treatment destabilizes the cytoplasmic membrane and thus changes the membrane tension, which immediately activates MS channels that are able to effectively transport aminoglycosides into the cytoplasm for downstream killing. Our findings reveal the biological effects of hypoionic shock on bacteria and can help to develop novel adjuvants for aminoglycoside potentiation to combat bacterial pathogens via activating MS channels.


Subject(s)
Aminoglycosides , Escherichia coli Proteins , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria , Escherichia coli , Escherichia coli Proteins/genetics , Ion Channels , Mice , Molecular Docking Simulation
12.
Acc Chem Res ; 54(3): 595-604, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33406830

ABSTRACT

Natural products with a high ratio of sp3-hybridized atoms and oxygen-substituted stereogenic centers represent privileged structures for the development of pharmaceuticals and chemical probes. The multiple oxygen functionalities of these natural products endow their potent and selective biological activities, although they significantly heighten the challenge of their chemical assemblies. We focused on developing efficient strategies for the total syntheses of this biologically and chemically important class of molecules. A convergent strategy is more advantageous than a linear strategy for designing a shorter synthetic route because a convergent strategy enables direct coupling of functionalized fragments whereas a linear strategy involves stepwise construction of a molecule from its terminus. Radical reactions are preferred over polar reactions for the coupling of heavily functionalized and sp3-rich fragments, as they allow for C(sp3)-C(sp3) coupling without damaging diverse polar functional groups. With these considerations in mind, we designed radical-based convergent strategies for assembling highly oxygenated natural products. Here we summarize the concise total syntheses of asimicin (1, antibiotic activity), 1-hydroxytaxinine (2, cytotoxicity), polyoxins (3, antifungal activity), and hikizimycin (4, anthelmintic activity) as representative examples. Retrosynthetic disconnection at the central part of these molecules produces highly substituted α-alkoxy radicals as synthons. In the synthetic direction, the α-alkoxy radicals were generated from the corresponding α-alkoxyacyl tellurides in a unified fashion, and then utilized for four distinct coupling reactions. Formation of the Et radical from Et3B and O2 homolytically cleaves the C-Te bond of α-alkoxyacyl telluride, and the facile expulsion of carbon monoxide from the acyl radical leads to the α-alkoxy radical. Dimerization of the stabilized α-alkoxy radical resulted in the core structure of 1 with 10 contiguous stereocenters. The coupling adduct was derivatized to 1 through the attachment of two different carbon chains (17 steps as the longest linear sequence). Alternatively, intermolecular addition reactions of the α-alkoxy radicals to electron-deficient C═C, C═N, and C═O bonds, followed by Et3B-mediated radical termination, led to the core structures of 2, 3, and 4, respectively. Intermolecular coupling between the α-alkoxy radical and the cyclohexenone derivative and intramolecular pinacol coupling gave rise to the 6/8/6-fused ring system of 2, which was transformed to 2 (26 steps). The two amino acid moieties of 3 were prepared by combining the α-alkoxy radical and the oxime and were then condensed to complete the synthesis of 3 (11 steps). Furthermore, a combination of α-alkoxyacyl telluride and Et3B/O2 realized a novel addition reaction of α-alkoxy radicals to aldehydes. This method was incorporated in the construction of the core 4-amino-5-deoxyundecose with 10 contiguous stereocenters, which was fabricated with two appendage structures to deliver 4. The four total syntheses described here demonstrate the versatility and robustness of intermolecular radical reactions. These syntheses will also provide new insights for retrosynthetic analyses in the field of organic chemistry and streamline synthetic routes to various bioactive natural products with multiple oxygen functionalities.


Subject(s)
Biological Products/chemical synthesis , Free Radicals/chemistry , Oxygen/chemistry , Aminoglycosides/chemistry , Biological Products/chemistry , Drug Design , Furans/chemical synthesis , Furans/chemistry , Pyrimidine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/chemistry , Quantum Theory , Stereoisomerism , Taxoids/chemical synthesis , Taxoids/chemistry
13.
Chemistry ; 28(36): e202200883, 2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35388562

ABSTRACT

The continuous emergence of antimicrobial resistance is causing a threat to patients infected by multidrug-resistant pathogens. In particular, the clinical use of aminoglycoside antibiotics, broad-spectrum antibacterials of last resort, is limited due to rising bacterial resistance. One of the major resistance mechanisms in Gram-positive and Gram-negative bacteria is phosphorylation of these amino sugars at the 3'-position by O-phosphotransferases [APH(3')s]. Structural alteration of these antibiotics at the 3'-position would be an obvious strategy to tackle this resistance mechanism. However, the access to such derivatives requires cumbersome multi-step synthesis, which is not appealing for pharma industry in this low-return-on-investment market. To overcome this obstacle and combat bacterial resistance mediated by APH(3')s, we introduce a novel regioselective modification of aminoglycosides in the 3'-position via palladium-catalyzed oxidation. To underline the effectiveness of our method for structural modification of aminoglycosides, we have developed two novel antibiotic candidates overcoming APH(3')s-mediated resistance employing only four synthetic steps.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria , Gram-Positive Bacteria , Humans , Phosphotransferases
14.
Bioorg Med Chem ; 73: 117011, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36191548

ABSTRACT

The total synthesis of capuramycin (1), which is a promising anti-tubercular antibiotics, has been accomplished using Ferrier-type I reaction as a key step. This total synthesis is an alternative approach to the synthesis of capuramycin and its analogues. The 3'-O-demethyl analogue (2), which exhibits an equivalent antibacterial activity as capuramycin (1) against Mycobacterium smegmatis and Mycobacterium avium, is suggested to have potential as a lead structure of capuramycin analogues because 2 is more accessible from a synthetic view point.


Subject(s)
Aminoglycosides , Mycobacterium smegmatis , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship
15.
Bioorg Chem ; 126: 105824, 2022 09.
Article in English | MEDLINE | ID: mdl-35636122

ABSTRACT

Despite their clinical importance, saving numerous human lifes, over- and mis-uses of antibiotics have created a strong selective pressure on bacteria, which induces the emergence of (multi)resistant strains. Antibioresistance is becoming so pregnant that since 2017, WHO lists bacteria threatening most human health (AWaRe, ESKAPE lists), and those for which new antibiotics are urgently needed. Since the century turn, this context is leading to a burst in the chemical synthesis of new antibiotics, mostly derived from natural antibiotics. Among them, aminoglycosides, and especially the neomycin family, exhibit broad spectrum of activity and remain clinically useful drugs. Therefore, numerous endeavours have been undertaken to modify aminoglycosides with the aim of overcoming bacterial resistances. After having replaced antibiotic discovery into an historical perspective, briefly surveyed the aminoglycoside mode of action and the associated resistance mechanisms, this review emphasized the chemical syntheses performed on the neomycin family and the corresponding structure activity relationships in order to reveal the really efficient modifications able to convert neomycin and its analogues into future drugs. This review would help researchers to strategically design novel aminoglycoside derivatives for the development of clinically viable drug candidates.


Subject(s)
Bacterial Infections , Neomycin , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemistry , Bacteria , Humans , Neomycin/chemistry , Neomycin/pharmacology , Paromomycin/chemistry , Paromomycin/pharmacology
16.
Molecules ; 27(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36364310

ABSTRACT

Chemical modification of old drugs is an important way to obtain new ones, and it has been widely used in developing new aminoglycoside antibiotics. However, many of the previous modifying strategies seem arbitrary for their lack of support from structural biological detail. In this paper, based on the structural information of aminoglycoside and its drug target, we firstly analyzed the reason that some 2'-N-acetylated products of aminoglycosides caused by aminoglycoside-modifying enzyme AAC(2') can partially retain activity, and then we designed, synthesized, and evaluated a series of 2'-modified kanamycin A derivatives. Bioassay results showed our modifying strategy was feasible. Our study provided valuable structure-activity relationship information, which would help researchers to develop new aminoglycoside antibiotics more effectively.


Subject(s)
Aminoglycosides , Kanamycin , Kanamycin/pharmacology , Kanamycin/chemistry , Aminoglycosides/chemistry , Anti-Bacterial Agents/chemistry , Structure-Activity Relationship , Biological Assay , Acetyltransferases
17.
Proteins ; 89(1): 94-106, 2021 01.
Article in English | MEDLINE | ID: mdl-32860271

ABSTRACT

Enhanced intracellular survival (Eis) proteins belonging to the superfamily of the GCN5-related N-acetyltransferases play important functions in mycobacterial pathogenesis. In Mycobacterium tuberculosis, Eis enhances the intracellular survival of the bacilli in macrophages by modulating the host immune response and is capable to chemically modify and inactivate aminoglycosides. In nontuberculous mycobacteria (NTM), Eis shares similar functions. However, Mycobacterium abscessus, a multidrug resistant NTM, possesses two functionally distinct Eis homologues, Eis1Mab and Eis2Mab . While Eis2Mab participates in virulence and aminoglycosides resistance, this is not the case for Eis1Mab, whose exact biological function remains to be determined. Herein, we show that overexpression of Eis1Mab in M. abscessus fails to induce resistance to aminoglycosides. To clarify why Eis1Mab is unable to modify this class of antibiotics, we solved its crystal structure bound to its cofactor, acetyl-CoA. The structure revealed that Eis1Mab has a typical homohexameric Eis-like organization. The structural analysis supported by biochemical approaches demonstrated that while Eis1Mab can acetylate small substrates, its active site is too narrow to accommodate aminoglycosides. Comparison with other Eis structures showed that an extended loop between strands 9 and 10 is blocking the access of large substrates to the active site and movement of helices 4 and 5 reduces the volume of the substrate-binding pocket to these compounds in Eis1Mab . Overall, this study underscores the molecular determinants explaining functional differences between Eis1Mab and Eis2Mab, especially those inherent to their capacity to modify aminoglycosides.


Subject(s)
Aminoglycosides , Mycobacterium abscessus , Acetyltransferases/chemistry , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Models, Molecular , Mycobacterium abscessus/metabolism
18.
J Am Chem Soc ; 143(13): 5038-5043, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33784078

ABSTRACT

AprD4 is a radical S-adenosyl-l-methionine (SAM) enzyme catalyzing C3'-deoxygenation of paromamine to form 4'-oxo-lividamine. It is the only 1,2-diol dehydratase in the radical SAM enzyme superfamily that has been identified and characterized in vitro. The AprD4 catalyzed 1,2-diol dehydration is a key step in the biosynthesis of several C3'-deoxy-aminoglycosides. While the regiochemistry of the hydrogen atom abstraction catalyzed by AprD4 has been established, the mechanism of the subsequent chemical transformation remains not fully understood. To investigate the mechanism, several substrate analogues were synthesized and their fates upon incubation with AprD4 were analyzed. The results support a mechanism involving formation of a ketyl radical intermediate followed by direct elimination of the C3'-hydroxyl group rather than that of a gem-diol intermediate generated via 1,2-migration of the C3'-hydroxyl group to C4'. The stereochemistry of hydrogen atom incorporation after radical-mediated dehydration was also established.


Subject(s)
Aminoglycosides/chemistry , Enzymes/chemistry , S-Adenosylmethionine/chemistry , Catalysis , Water/chemistry
19.
Chembiochem ; 22(9): 1563-1567, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33410196

ABSTRACT

We report the characterization of amphiphilic aminoglycoside conjugates containing luminophores with aggregation-induced emission properties as transfection reagents. These inherently luminescent transfection vectors are capable of binding plasmid DNA through electrostatic interactions; this binding results in an emission "on" signal due to restriction of intramolecular motion of the luminophore core. The luminescent cationic amphiphiles effectively transferred plasmid DNA into mammalian cells (HeLa, HEK 293T), as proven by expression of a red fluorescent protein marker. The morphologies of the aggregates were investigated by microscopy as well as ζ-potential and dynamic light-scattering measurements. The transfection efficiencies using luminescent cationic amphiphiles were similar to that of the gold-standard transfection reagent Lipofectamine® 2000.


Subject(s)
Aminoglycosides/chemistry , Transfection/methods , Aminoglycosides/pharmacology , Animals , Cell Survival/drug effects , HEK293 Cells , HeLa Cells , Humans , Lipids/chemistry , Microscopy, Confocal , Plasmids/chemistry , Plasmids/metabolism , Static Electricity , Tobramycin/chemistry , Tobramycin/pharmacology
20.
RNA ; 25(2): 247-254, 2019 02.
Article in English | MEDLINE | ID: mdl-30413565

ABSTRACT

Aminoglycosides containing a 2-deoxystreptamine core (AGs) represent a large family of antibiotics that target the ribosome. These compounds promote miscoding, inhibit translocation, and inhibit ribosome recycling. AG binding to helix h44 of the small subunit induces rearrangement of A-site nucleotides A1492 and A1493, which promotes a key open-to-closed conformational change of the subunit and thereby increases miscoding. Mechanisms by which AGs inhibit translocation and recycling remain less clear. Structural studies have revealed a secondary AG binding site in H69 of the large subunit, and it has been proposed that interaction at this site is crucial for inhibition of translocation and recycling. Here, we analyze ribosomes with mutations targeting either or both AG binding sites. Assaying translocation, we find that ablation of the h44 site increases the IC50 values for AGs dramatically, while removal of the H69 site increases these values modestly. This suggests that the AG-h44 interaction is primarily responsible for inhibition, with H69 playing a minor role. Assaying recycling, we find that mutation of h44 has no effect on AG inhibition, consistent with a primary role for AG-H69 interaction. Collectively, these findings help clarify the roles of the two AG binding sites in mechanisms of inhibition by these compounds.


Subject(s)
Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects , Translocation, Genetic/drug effects , Bacterial Infections/drug therapy , Binding Sites/genetics , Hexosamines/chemistry , Protein Structure, Secondary/genetics , Protein Synthesis Inhibitors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL