Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
RNA ; 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33262249

ABSTRACT

We have recently reported on an experimental model of mitochondrial mistranslation conferred by amino acid exchange V338Y in the mitochondrial ribosomal protein MrpS5. Here we used a combination of RNA-Seq and metabolic profiling of homozygous transgenic MrpS5V338Y/V338Y mice to analyze the changes associated with the V338Y mutation in post-mitotic skeletal muscle. Metabolic profiling demonstrated age-dependent metabolic changes in the mutant V338Y animals, which included enhanced levels of age-associated metabolites and which were accompanied by increased glycolysis, lipid desaturation and eicosanoid biosynthesis, and alterations of the pentose phosphate pathway. In addition, transcriptome signatures of aged V338Y mutant muscle pointed to elevated inflammation, likely reflecting the increased levels of bioactive lipids. Our findings indicate that mistranslation-mediated chronic impairment of mitochondrial function affects specific bioenergetic processes in muscle in an age-dependent manner.

2.
Commun Biol ; 2: 381, 2019.
Article in English | MEDLINE | ID: mdl-31637312

ABSTRACT

Translation fidelity is the limiting factor in the accuracy of gene expression. With an estimated frequency of 10-4, errors in mRNA decoding occur in a mostly stochastic manner. Little is known about the response of higher eukaryotes to chronic loss of ribosomal accuracy as per an increase in the random error rate of mRNA decoding. Here, we present a global and comprehensive picture of the cellular changes in response to translational accuracy in mammalian ribosomes impaired by genetic manipulation. In addition to affecting established protein quality control pathways, such as elevated transcript levels for cytosolic chaperones, activation of the ubiquitin-proteasome system, and translational slowdown, ribosomal mistranslation led to unexpected responses. In particular, we observed increased mitochondrial biogenesis associated with import of misfolded proteins into the mitochondria and silencing of the unfolded protein response in the endoplasmic reticulum.


Subject(s)
Organelle Biogenesis , Ribosomes/genetics , Ribosomes/metabolism , Unfolded Protein Response/genetics , Amino Acid Substitution , Endoplasmic Reticulum/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling , HEK293 Cells , Humans , Mitochondria/metabolism , Mutation , Protein Biosynthesis , Protein Transport/genetics , Proteostasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
3.
EMBO Rep ; 19(11)2018 11.
Article in English | MEDLINE | ID: mdl-30237157

ABSTRACT

The 1555 A to G substitution in mitochondrial 12S A-site rRNA is associated with maternally transmitted deafness of variable penetrance in the absence of otherwise overt disease. Here, we recapitulate the suggested A1555G-mediated pathomechanism in an experimental model of mitoribosomal mistranslation by directed mutagenesis of mitoribosomal protein MRPS5. We first establish that the ratio of cysteine/methionine incorporation and read-through of mtDNA-encoded MT-CO1 protein constitute reliable measures of mitoribosomal misreading. Next, we demonstrate that human HEK293 cells expressing mutant V336Y MRPS5 show increased mitoribosomal mistranslation. As for immortalized lymphocytes of individuals with the pathogenic A1555G mutation, we find little changes in the transcriptome of mutant V336Y MRPS5 HEK cells, except for a coordinated upregulation of transcripts for cytoplasmic ribosomal proteins. Homozygous knock-in mutant Mrps5 V338Y mice show impaired mitochondrial function and a phenotype composed of enhanced susceptibility to noise-induced hearing damage and anxiety-related behavioral alterations. The experimental data in V338Y mutant mice point to a key role of mitochondrial translation and function in stress-related behavioral and physiological adaptations.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/genetics , Ribosomal Proteins/genetics , Aging/genetics , Animals , Behavior, Animal , Brain/cytology , Cysteine/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Escherichia coli Proteins/genetics , HEK293 Cells , Hearing Disorders/genetics , Humans , Methionine/metabolism , Mice, Transgenic , Mitochondria/genetics , Noise/adverse effects , Protein Biosynthesis , RNA, Messenger , Ribosomes/genetics , Ribosomes/metabolism , Stress, Physiological/genetics
4.
Sci Transl Med ; 7(288): 288ra75, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995221

ABSTRACT

The antibiotic spectinomycin is a potent inhibitor of bacterial protein synthesis with a unique mechanism of action and an excellent safety index, but it lacks antibacterial activity against most clinically important pathogens. A series of N-benzyl-substituted 3'-(R)-3'-aminomethyl-3'-hydroxy spectinomycins was developed on the basis of a computational analysis of the aminomethyl spectinomycin binding site and structure-guided synthesis. These compounds had ribosomal inhibition values comparable to spectinomycin but showed increased potency against the common respiratory tract pathogens Streptococcus pneumoniae, Haemophilus influenzae, Legionella pneumophila, and Moraxella catarrhalis, as well as the sexually transmitted bacteria Neisseria gonorrhoeae and Chlamydia trachomatis. Non-ribosome-binding 3'-(S) isomers of the lead compounds demonstrated weak inhibitory activity in in vitro protein translation assays and poor antibacterial activity, indicating that the antibacterial activity of the series remains on target against the ribosome. Compounds also demonstrated no mammalian cytotoxicity, improved microsomal stability, and favorable pharmacokinetic properties in rats. The lead compound from the series exhibited excellent chemical stability superior to spectinomycin; no interaction with a panel of human receptors and drug metabolism enzymes, suggesting low potential for adverse reactions or drug-drug interactions in vivo; activity in vitro against a panel of penicillin-, macrolide-, and cephalosporin-resistant S. pneumoniae clinical isolates; and the ability to cure mice of fatal pneumococcal pneumonia and sepsis at a dose of 5 mg/kg. Together, these studies indicate that N-benzyl aminomethyl spectinomycins are suitable for further development to treat drug-resistant respiratory tract and sexually transmitted bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Discovery , Drug Resistance, Bacterial , Respiratory Tract Infections/drug therapy , Sexually Transmitted Diseases, Bacterial/drug therapy , Spectinomycin/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacokinetics , Bacteria/metabolism , Bacteria/pathogenicity , Bacterial Proteins/biosynthesis , Chlorocebus aethiops , Computer Simulation , Computer-Aided Design , Disease Models, Animal , Drug Interactions , Drug Stability , Humans , Male , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/microbiology , Ribosomes/drug effects , Ribosomes/metabolism , Sexually Transmitted Diseases, Bacterial/diagnosis , Sexually Transmitted Diseases, Bacterial/microbiology , Spectinomycin/adverse effects , Spectinomycin/analogs & derivatives , Spectinomycin/chemical synthesis , Spectinomycin/pharmacokinetics , Structure-Activity Relationship , Vero Cells
5.
mBio ; 5(5): e01827-14, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25271289

ABSTRACT

UNLABELLED: The emerging epidemic of drug resistance places the development of efficacious and safe antibiotics in the spotlight of current research. Here, we report the design of next-generation aminoglycosides. Discovery efforts were driven by rational synthesis focusing on 4' alkylations of the aminoglycoside paromomycin, with the goal to alleviate the most severe and disabling side effect of aminoglycosides-irreversible hearing loss. Compounds were evaluated for target activity in in vitro ribosomal translation assays, antibacterial potency against selected pathogens, cytotoxicity against mammalian cells, and in vivo ototoxicity. The results of this study produced potent compounds with excellent selectivity at the ribosomal target, promising antibacterial activity, and little, if any, ototoxicity upon chronic administration. The favorable biocompatibility profile combined with the promising antibacterial activity emphasizes the potential of next-generation aminoglycosides in the treatment of infectious diseases without the risk of ototoxicity. IMPORTANCE: The ever-widening epidemic of multidrug-resistant infectious diseases and the paucity of novel antibacterial agents emerging from modern screening platforms mandate the reinvestigation of established drugs with an emphasis on improved biocompatibility and overcoming resistance mechanisms. Here, we describe the preparation and evaluation of derivatives of the established aminoglycoside antibiotic paromomycin that effectively remove its biggest deficiency, ototoxicity, and overcome certain bacterial resistance mechanisms.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Aminoglycosides/chemical synthesis , Animals , Anti-Bacterial Agents/chemical synthesis , Bacterial Infections/drug therapy , Escherichia coli/drug effects , Guinea Pigs , Hexosamines/chemical synthesis , Hexosamines/pharmacology , Male , Mice , Mice, Inbred ICR , Microbial Sensitivity Tests , NIH 3T3 Cells , Phylogeny , RNA, Ribosomal, 16S/genetics , Ribosomes/drug effects , Staphylococcus aureus/drug effects
6.
ChemMedChem ; 9(9): 2074-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25045149

ABSTRACT

A series of apramycin derivatives was prepared and investigated for antibacterial activity and the ability to inhibit protein synthesis in cell-free translation assays. The effect of various modifications at the 6'- and N7'-positions on antiribosomal activity is discussed in terms of their influence on drug binding to specific residues in the decoding A-site. These studies contribute to the development of a structure-activity relationship for the antibacterial activity of the apramycin class of aminoglycosides and to the future design and development of more active and less toxic antibiotics.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Nebramycin/analogs & derivatives , Protein Synthesis Inhibitors/chemical synthesis , Protein Synthesis Inhibitors/pharmacology , Animals , Bacteria/drug effects , Carbohydrate Sequence , Catalytic Domain/drug effects , Cell-Free System , Molecular Sequence Data , Nebramycin/chemical synthesis , Nebramycin/pharmacology , Protein Biosynthesis/drug effects , Rabbits , Ribosomes/drug effects , Structure-Activity Relationship
7.
Nat Commun ; 5: 3112, 2014.
Article in English | MEDLINE | ID: mdl-24473108

ABSTRACT

Clinical use of 2-deoxystreptamine aminoglycoside antibiotics, which target the bacterial ribosome, is compromised by adverse effects related to limited drug selectivity. Here we present a series of 4',6'-O-acetal and 4'-O-ether modifications on glucopyranosyl ring I of aminoglycosides. Chemical modifications were guided by measuring interactions between the compounds synthesized and ribosomes harbouring single point mutations in the drug-binding site, resulting in aminoglycosides that interact poorly with the drug-binding pocket of eukaryotic mitochondrial or cytosolic ribosomes. Yet, these compounds largely retain their inhibitory activity for bacterial ribosomes and show antibacterial activity. Our data indicate that 4'-O-substituted aminoglycosides possess increased selectivity towards bacterial ribosomes and little activity for any of the human drug-binding pockets.


Subject(s)
Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Aminoglycosides/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Base Sequence , Cell-Free System , Crystallography, X-Ray , Disease Models, Animal , Drug Interactions , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Humans , Inhibitory Concentration 50 , Male , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Mycobacterium smegmatis/drug effects , Nucleic Acid Conformation , Protein Biosynthesis/drug effects , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , Ribosomes/metabolism , Sepsis/drug therapy , Staphylococcus aureus/drug effects
8.
Antimicrob Agents Chemother ; 56(12): 6104-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22948879

ABSTRACT

The kanamycins form an important subgroup of the 4,6-disubstituted 2-deoxystreptamine aminoglycoside antibiotics, comprising kanamycin A, kanamycin B, tobramycin, and dibekacin. These compounds interfere with protein synthesis by targeting the ribosomal decoding A site, and they differ in the numbers and locations of amino and hydroxy groups of the glucopyranosyl moiety (ring I). We synthesized kanamycin analogues characterized by subtle variations of the 2' and 6' substituents of ring I. The functional activities of the kanamycins and the synthesized analogues were investigated (i) in cell-free translation assays on wild-type and mutant bacterial ribosomes to study drug-target interaction, (ii) in MIC assays to assess antibacterial activity, and (iii) in rabbit reticulocyte translation assays to determine activity on eukaryotic ribosomes. Position 2' forms an intramolecular H bond with O5 of ring II, helping the relative orientations of the two rings with respect to each other. This bond becomes critical for drug activity when a 6'-OH substituent is present.


Subject(s)
Anti-Bacterial Agents/pharmacology , Kanamycin/analogs & derivatives , Kanamycin/pharmacology , Amines/chemistry , Animals , Anti-Bacterial Agents/chemistry , Carbohydrate Sequence , Hydroxylation , Kanamycin/chemistry , Luciferases/chemistry , Luciferases/genetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Mutation , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , RNA, Bacterial/genetics , RNA, Ribosomal/genetics , Rabbits , Reticulocytes/drug effects , Ribosomes/drug effects , Structure-Activity Relationship
9.
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
10.
Antimicrob Agents Chemother ; 55(10): 4712-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21768509

ABSTRACT

Capreomycin and the structurally similar compound viomycin are cyclic peptide antibiotics which are particularly active against Mycobacterium tuberculosis, including multidrug resistant strains. Both antibiotics bind across the ribosomal interface involving 23S rRNA helix 69 (H69) and 16S rRNA helix 44 (h44). The binding site of tuberactinomycins in h44 partially overlaps with that of aminoglycosides, and they share with these drugs the side effect of irreversible hearing loss. Here we studied the drug target interaction on ribosomes modified by site-directed mutagenesis. We identified rRNA residues in h44 as the main determinants of phylogenetic selectivity, predict compensatory evolution to impact future resistance development, and propose mechanisms involved in tuberactinomycin ototoxicity, which may enable the development of improved, less-toxic derivatives.


Subject(s)
Antitubercular Agents/pharmacology , Capreomycin/pharmacology , Mycobacterium tuberculosis/drug effects , Ribosomes/drug effects , Viomycin/pharmacology , Aminoglycosides/pharmacology , Antitubercular Agents/metabolism , Antitubercular Agents/toxicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capreomycin/metabolism , Capreomycin/toxicity , Drug Resistance, Multiple, Bacterial/genetics , Enviomycin/analogs & derivatives , Enviomycin/pharmacology , Enviomycin/toxicity , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , RNA, Ribosomal, 16S/metabolism , RNA, Ribosomal, 23S/metabolism , Viomycin/metabolism , Viomycin/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...