Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Int J Mol Sci ; 23(8)2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35457201

ABSTRACT

We have recently identified point mutation V336Y in mitoribosomal protein Mrps5 (uS5m) as a mitoribosomal ram (ribosomal ambiguity) mutation conferring error-prone mitochondrial protein synthesis. In vivo in transgenic knock-in animals, homologous mutation V338Y was associated with a discrete phenotype including impaired mitochondrial function, anxiety-related behavioral alterations, enhanced susceptibility to noise-induced hearing damage, and accelerated metabolic aging in muscle. To challenge the postulated link between Mrps5 V338Y-mediated misreading and the in vivo phenotype, we introduced mutation G315R into the mouse Mrps5 gene as Mrps5 G315R is homologous to the established bacterial ram mutation RpsE (uS5) G104R. However, in contrast to bacterial translation, the homologous G → R mutation in mitoribosomal Mrps5 did not affect the accuracy of mitochondrial protein synthesis. Importantly, in the absence of mitochondrial misreading, homozygous mutant MrpS5G315R/G315R mice did not show a phenotype distinct from wild-type animals.


Subject(s)
Mitochondrial Proteins , Ribosomal Proteins , Animals , Mice , Mitochondrial Proteins/genetics , Mutation , Phenotype , Phylogeny , Protein Biosynthesis , Ribosomal Proteins/genetics
2.
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
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.
J Am Chem Soc ; 141(12): 5051-5061, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30793894

ABSTRACT

Infectious diseases due to multidrug-resistant pathogens, particularly carbapenem-resistant Enterobacteriaceae (CREs), present a major and growing threat to human health and society, providing an urgent need for the development of improved potent antibiotics for their treatment. We describe the design and development of a new class of aminoglycoside antibiotics culminating in the discovery of propylamycin. Propylamycin is a 4'-deoxy-4'-alkyl paromomycin whose alkyl substituent conveys excellent activity against a broad spectrum of ESKAPE pathogens and other Gram-negative infections, including CREs, in the presence of numerous common resistance determinants, be they aminoglycoside modifying enzymes or rRNA methyl transferases. Importantly, propylamycin is demonstrated not to be susceptible to the action of the ArmA resistance determinant whose presence severely compromises the action of plazomicin and all other 4,6-disubstituted 2-deoxystreptamine aminoglycosides. The lack of susceptibility to ArmA, which is frequently encoded on the same plasmid as carbapenemase genes, ensures that propylamycin will not suffer from problems of cross-resistance when used in combination with carbapenems. Cell-free translation assays, quantitative ribosome footprinting, and X-ray crystallography support a model in which propylamycin functions by interference with bacterial protein synthesis. Cell-free translation assays with humanized bacterial ribosomes were used to optimize the selectivity of propylamycin, resulting in reduced ototoxicity in guinea pigs. In mouse thigh and septicemia models of Escherichia coli, propylamycin shows excellent efficacy, which is better than paromomycin. Overall, a simple novel deoxy alkyl modification of a readily available aminoglycoside antibiotic increases the inherent antibacterial activity, effectively combats multiple mechanisms of aminoglycoside resistance, and minimizes one of the major side effects of aminoglycoside therapy.


Subject(s)
Aminoglycosides/chemical synthesis , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Drug Design , Drug Resistance, Bacterial/drug effects , Enterobacteriaceae/drug effects , Aminoglycosides/chemistry , Animals , Anti-Bacterial Agents/chemistry , Chemistry Techniques, Synthetic , Guinea Pigs , Hexosamines/chemical synthesis , Hexosamines/chemistry , Hexosamines/pharmacology , Hexosamines/toxicity , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Structure-Activity Relationship
5.
J Toxicol Environ Health A ; 81(20): 1041-1057, 2018.
Article in English | MEDLINE | ID: mdl-30239325

ABSTRACT

Exposure to heavy metals may lead to hearing impairment. However, experimental studies have not explored this issue with and without noise exposure in mature animals with environmentally relevant doses. The aim of this study was to investigate ototoxicity produced by lead (Pb) and cadmium (Cd) and noise, singly and in combination, in the adult CBA/CaJ mouse. Metals were delivered via drinking water (0.03 mM, 1 mM, and 3 mM Pb; or 30, 100, and 300 µM Cd) for 12 weeks, resulting in environmentally- and occupationally relevant mean (± standard deviations) blood levels of Pb (2.89 ± 0.44, 38.5 ± 4.9, and 60.1 ± 6.6 µg/dl, respectively) and Cd (1.3 ± 0.23, 6.37 ± 0.87, 27.2 ± 4.1 µg/L, respectively). Metal treatment was also combined with a noise exposure consisting of a 105 dB broadband (2-20 kHz) stimulus for 2 hr or a sham exposure. Auditory performance was determined by comparing auditory brainstem responses (ABR) and distortion product otoacoustic emissions (DPOAE) at baseline and after 11 weeks of metal treatment. Metal-exposed animals did not develop significant auditory deficits and did not exhibit morphological damage to cochlear hair cells. In contrast, noise-exposed animals, including those exposed to combinations of metals and noise, demonstrated significant hair cell loss, reduced DPOAE amplitudes, and ABR threshold shifts of 42.2 ± 13 dB at 32 kHz (105 dB noise alone). No significant potentiation or synergistic effects were found in groups exposed to multiple agents. This study establishes a highly reproducible adult mouse model that may be used to evaluate a variety of environmental exposure mixtures.


Subject(s)
Auditory Threshold/drug effects , Cadmium/adverse effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss, Noise-Induced/etiology , Lead/adverse effects , Noise/adverse effects , Otoacoustic Emissions, Spontaneous/drug effects , Animals , Environmental Pollutants/toxicity , Hearing Loss, Noise-Induced/chemically induced , Male , Mice , Mice, Inbred CBA
6.
Nucleic Acids Res ; 43(17): 8601-13, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26264664

ABSTRACT

Leishmaniasis comprises an array of diseases caused by pathogenic species of Leishmania, resulting in a spectrum of mild to life-threatening pathologies. Currently available therapies for leishmaniasis include a limited selection of drugs. This coupled with the rather fast emergence of parasite resistance, presents a dire public health concern. Paromomycin (PAR), a broad-spectrum aminoglycoside antibiotic, has been shown in recent years to be highly efficient in treating visceral leishmaniasis (VL)-the life-threatening form of the disease. While much focus has been given to exploration of PAR activities in bacteria, its mechanism of action in Leishmania has received relatively little scrutiny and has yet to be fully deciphered. In the present study we present an X-ray structure of PAR bound to rRNA model mimicking its leishmanial binding target, the ribosomal A-site. We also evaluate PAR inhibitory actions on leishmanial growth and ribosome function, as well as effects on auditory sensory cells, by comparing several structurally related natural and synthetic aminoglycoside derivatives. The results provide insights into the structural elements important for aminoglycoside inhibitory activities and selectivity for leishmanial cytosolic ribosomes, highlighting a novel synthetic derivative, compound 3: , as a prospective therapeutic candidate for the treatment of VL.


Subject(s)
Antiprotozoal Agents/chemistry , Leishmania/drug effects , Paromomycin/chemistry , Protein Synthesis Inhibitors/chemistry , Ribosomes/drug effects , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/toxicity , Binding Sites , Cell Line , Computer Simulation , Guinea Pigs , Humans , Leishmania/growth & development , Macrophages/parasitology , Male , Models, Molecular , Neomycin/analogs & derivatives , Neomycin/chemistry , Neomycin/toxicity , Paromomycin/pharmacology , Paromomycin/toxicity , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/toxicity , RNA, Ribosomal/chemistry , Ribosomes/chemistry
7.
Environ Toxicol ; 32(3): 869-876, 2017 03.
Article in English | MEDLINE | ID: mdl-27257108

ABSTRACT

OBJECTIVES: The vestibular system allows the perception of position and motion and its dysfunction presents as motion impairment, vertigo and balance abnormalities, leading to debilitating psychological discomfort and difficulty performing daily tasks. Although declines and deficits in vestibular function have been noted in rats exposed to lead (Pb) and in humans exposed to Pb and cadmium (Cd), no studies have directly examined the pathological and pathophysiological effects upon the vestibular apparatus of the inner ear. METHODS: Eighteen young adult mice were exposed through their drinking water (3 mM Pb, 300 µM Cd, or a control treatment) for 10 weeks. Before and after treatment, they underwent a vestibular assessment, consisting of a rotarod performance test and a novel head stability test to measure the vestibulocolic reflex. At the conclusion of the study, the utricles were analyzed immunohistologically for condition of hair cells and nerve fibers. RESULTS: Increased levels of Pb exposure correlated with decreased head stability in space; no significant decline in performance on rotarod test was found. No damage to the hair cells or the nerve fibers of the utricle was observed in histology. CONCLUSIONS: The young adult CBA/CaJ mouse is able to tolerate occupationally-relevant Pb and Cd exposure well, but the correlation between Pb exposure and reduced head stability suggests that Pb exposure causes a decline in vestibular function. © 2016 Wiley Periodicals, Inc. Environ Toxicol, 2016. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 869-876, 2017.


Subject(s)
Brain/drug effects , Cadmium/toxicity , Lead/toxicity , Animals , Bone and Bones/chemistry , Bone and Bones/metabolism , Brain/metabolism , Brain/pathology , Drinking Water/chemistry , Male , Mice , Mice, Inbred CBA
8.
J Biol Chem ; 289(4): 2318-30, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24302717

ABSTRACT

There is compelling evidence that aminoglycoside (AG) antibiotics can induce the mammalian ribosome to suppress disease-causing nonsense mutations and partially restore the expression of functional proteins. However, prolonged AG treatment can cause detrimental side effects in patients, including most prominently, ototoxicity. Recent mechanistic discussions have considered the relative contributions of mitochondrial and cytoplasmic protein synthesis inhibition to AG-induced ototoxicity. We show that AGs inhibit mitochondrial protein synthesis in mammalian cells and perturb cell respiration, leading to a time- and dose-dependent increase in superoxide overproduction and accumulation of free ferrous iron in mitochondria caused by oxidative damage of mitochondrial aconitase, ultimately leading to cell apoptosis via the Fenton reaction. These deleterious effects increase with the increased potency of AG to inhibit the mitochondrial rather than cytoplasmic protein synthesis, which in turn correlates with their ototoxic potential in both murine cochlear explants and the guinea pig in vivo. The deleterious effects of AGs were alleviated in synthetic derivatives specially designed for the treatment of genetic diseases caused by nonsense mutations and possessing low affinity toward mitochondrial ribosomes. This work highlights the benefit of a mechanism-based drug redesign strategy that can maximize the translational value of "readthrough therapy" while mitigating drug-induced side effects. This approach holds promise for patients suffering from genetic diseases caused by nonsense mutations.


Subject(s)
Aminoglycosides/pharmacology , Cytoplasm/metabolism , Mitochondria/metabolism , Protein Synthesis Inhibitors/pharmacology , Ribosomes/metabolism , Aminoglycosides/adverse effects , Animals , Apoptosis/drug effects , Cochlea/metabolism , Dose-Response Relationship, Drug , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Guinea Pigs , HeLa Cells , Humans , Mice , Mitochondrial Proteins/biosynthesis , Oxygen Consumption/drug effects , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/adverse effects , Reactive Oxygen Species/metabolism
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.
Am J Respir Cell Mol Biol ; 50(4): 805-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24251786

ABSTRACT

New drugs are needed to enhance premature termination codon (PTC) suppression to treat the underlying cause of cystic fibrosis (CF) and other diseases caused by nonsense mutations. We tested new synthetic aminoglycoside derivatives expressly developed for PTC suppression in a series of complementary CF models. Using a dual-luciferase reporter system containing the four most prevalent CF transmembrane conductance regulator (CFTR) nonsense mutations (G542X, R553X, R1162X, and W1282X) within their local sequence contexts (the three codons on either side of the PTC), we found that NB124 promoted the most readthrough of G542X, R1162X, and W1282X PTCs. NB124 also restored full-length CFTR expression and chloride transport in Fischer rat thyroid cells stably transduced with a CFTR-G542XcDNA transgene, and was superior to gentamicin and other aminoglycosides tested. NB124 restored CFTR function to roughly 7% of wild-type activity in primary human bronchial epithelial (HBE) CF cells (G542X/delF508), a highly relevant preclinical model with endogenous CFTR expression. Efficacy was further enhanced by addition of the CFTR potentiator, ivacaftor (VX-770), to airway cells expressing CFTR PTCs. NB124 treatment rescued CFTR function in a CF mouse model expressing a human CFTR-G542X transgene; efficacy was superior to gentamicin and exhibited favorable pharmacokinetic properties, suggesting that in vitro results translated to clinical benefit in vivo. NB124 was also less cytotoxic than gentamicin in a tissue-based model for ototoxicity. These results provide evidence that NB124 and other synthetic aminoglycosides provide a 10-fold improvement in therapeutic index over gentamicin and other first-generation aminoglycosides, providing a promising treatment for a wide array of CFTR nonsense mutations.


Subject(s)
Aminoglycosides/pharmacology , Aminophenols/pharmacology , Codon, Nonsense/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/drug therapy , Quinolones/pharmacology , Aminoglycosides/chemical synthesis , Aminoglycosides/pharmacokinetics , Aminoglycosides/toxicity , Aminophenols/pharmacokinetics , Animals , Biological Transport , Cell Line , Chlorides/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Drug Synergism , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred CFTR , Mice, Transgenic , Organ of Corti/drug effects , Organ of Corti/pathology , Quinolones/pharmacokinetics , Rats , Rats, Inbred F344 , Time Factors , Transfection
11.
Expert Opin Emerg Drugs ; 16(2): 235-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21247358

ABSTRACT

INTRODUCTION: Approximately 5% of the population worldwide suffers from industrial, military or recreational noise-induced hearing loss (NIHL) at a great economic cost and detriment to the quality of life of the affected individuals. This review discusses pharmacological strategies to attenuate NIHL that have been developed in animal models and that are now beginning to be tested in field trials. AREAS COVERED: The review describes the epidemiology, pathology and pathophysiology of NIHL in experimental animals and humans. The underlying molecular mechanisms of damage are then discussed as a basis for therapeutic approaches to ameliorate the loss of auditory function. Finally, studies in military, industrial and recreational settings are evaluated. Literature was searched using the terms 'noise-induced hearing loss' and 'noise trauma'. EXPERT OPINION: NIHL, in principle, can be prevented. With the current pace of development, oral drugs to protect against NIHL should be available within the next 5-10 years. Positive results from ongoing trials combined with additional laboratory tests might accelerate the time from the bench to clinical treatment.


Subject(s)
Hearing Loss, Noise-Induced/drug therapy , Animals , Clinical Trials as Topic , Humans
12.
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
13.
J Neurochem ; 108(5): 1226-36, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141081

ABSTRACT

Post-translational modification of histones is an important form of chromatin regulation impacting transcriptional activation. Histone acetyltransferases, for example, acetylate lysine residues on histone tails thereby enhancing gene transcription, while histone deacetylases (HDACs) remove those acetyl groups and repress gene transcription. Deficient histone acetylation is associated with pathologies, and histone deacetylase inhibitors have been studied in the treatment of cancer and neurodegenerative diseases. Here we explore histone acetylation in cochlear sensory cells following a challenge with gentamicin, an aminoglycoside antibiotic known to cause loss of auditory hair cells and hearing. The addition of the drug to organotypic cultures of the mouse organ of Corti decreased the acetylation of histone core proteins (H2A Ack5, H2B Ack12, H3 Ack9, and H4 Ack8) followed by a loss of sensory cells. Protein levels of HDAC1, HDAC3 and HDAC4 were increased while the histone acetyltransferases such as CREB-binding protein and p300 remained unchanged. We next hypothesized that protecting histone acetylation should prevent cell death and tested the effects of HDAC-inhibitors on the actions of gentamicin. Co-treatment with trichostatin A maintained near-normal levels of acetylation of histone core proteins in cochlear hair cells and attenuated gentamicin-induced cell death. The addition of sodium butyrate also rescued hair cells from damage by gentamicin. The results are consistent with an involvement of deficient histone acetylation in aminoglycoside-induced hair cell death and point to the potential value of HDAC-inhibitors in protection from the side effects of these drugs.


Subject(s)
Gentamicins/pharmacology , Hair Cells, Auditory/drug effects , Histones/metabolism , Protein Synthesis Inhibitors/pharmacology , Acetylation/drug effects , Animals , Cell Count/methods , Cell Death/drug effects , Cochlea/cytology , Dose-Response Relationship, Drug , Drug Interactions , Female , Gene Expression Regulation/drug effects , Hair Cells, Auditory/cytology , Hydroxamic Acids/pharmacology , Male , Mice , Mice, Inbred CBA , Organ Culture Techniques , Time Factors
14.
Sci Rep ; 9(1): 9273, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31239523

ABSTRACT

Animal-based studies have provided important insights into the structural and functional consequences of noise exposure on the cochlea. Yet, less is known about the molecular mechanisms by which noise induces cochlear damage, particularly at relatively low exposure levels. While there is ample evidence that noise exposure leads to changes in inner ear metabolism, the specific effects of noise exposure on the cochlear metabolome are poorly understood. In this study we applied liquid chromatography-coupled tandem mass spectrometry (LC-MS/MS)-based metabolomics to analyze the effects of noise on the mouse inner ear. Mice were exposed to noise that induces temporary threshold shifts, synaptopathy and permanent hidden hearing loss. Inner ears were harvested immediately after exposure and analyzed by targeted metabolomics for the relative abundance of 220 metabolites across the major metabolic pathways in central carbon metabolism. We identified 40 metabolites differentially affected by noise. Our approach detected novel noise-modulated metabolites and pathways, as well as some already linked to noise exposure or cochlear function such as neurotransmission and oxidative stress. Furthermore, it showed that metabolic effects of noise on the inner ear depend on the intensity and duration of exposure. Collectively, our results illustrate that metabolomics provides a powerful approach for the characterization of inner ear metabolites affected by auditory trauma. This type of information could lead to the identification of drug targets and novel therapies for noise-induced hearing loss.


Subject(s)
Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/metabolism , Metabolome , Noise/adverse effects , Animals , Auditory Threshold , Ear, Inner/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/pathology , Mice , Mice, Inbred CBA , Tandem Mass Spectrometry
15.
Hear Res ; 243(1-2): 87-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18573325

ABSTRACT

Commercially obtained aged male CBA/J mice presented a complex pattern of hearing loss and morphological changes. A significant threshold shift in auditory brainstem responses (ABR) occurred at 3 months of age at 4 kHz without apparent loss of hair cells, rising slowly at later ages accompanied by loss of apical hair cells. A delayed high-frequency deficit started at 24 kHz around the age of 12 months. At 20-26 months, threshold shifts at 12 and 24 kHz and the accompanying hair cell loss at the base of the cochlea were highly variable with some animals appearing almost normal and others showing large deficits. Spiral ganglion cells degenerated by 18 months in all regions of the cochlea, with cell density reduced by approximately 25%. There was no degeneration of the stria vascularis and the endocochlear potential remained stable from 3 to 25 months of age regardless of whether the animals had normal or highly elevated ABR thresholds. The slow high-frequency hearing loss combined with a modest reduction of ganglion cell density and an unchanged endocochlear potential suggest sensorineural presbycusis. The superimposed early hearing loss at low frequencies, which is not seen in animals bred in-house, may complicate the use of these animals as a presbycusis model.


Subject(s)
Aging/pathology , Presbycusis/pathology , Animals , Auditory Threshold , Cochlea/pathology , Cochlear Microphonic Potentials , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory/pathology , Hearing Loss, High-Frequency/etiology , Hearing Loss, High-Frequency/pathology , Hearing Loss, High-Frequency/physiopathology , Male , Mice , Mice, Inbred CBA , Presbycusis/etiology , Presbycusis/physiopathology , Spiral Ganglion/pathology , Stria Vascularis/pathology
16.
ACS Infect Dis ; 4(7): 1114-1120, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29708331

ABSTRACT

Syntheses of the 6'- N-(2-hydroxyethyl) and 1- N-(4-amino-2 S-hydroxybutyryl) derivatives of the 4,6-aminoglycoside sisomicin and that of the doubly modified 1- N-(4-amino-2 S-hydroxybutyryl)-6'- N-(2-hydroxyethyl) derivative known as plazomicin are reported together with their antibacterial and antiribosomal activities and selectivities. The 6'- N-(2-hydroxyethyl) modification results in a moderate increase in prokaryotic/eukaryotic ribosomal selectivity, whereas the 1- N-(4-amino-2 S-hydroxybutyryl) modification has the opposite effect. When combined in plazomicin, the effects of the two groups on ribosomal selectivity cancel each other out, leading to the prediction that plazomicin will exhibit ototoxicity comparable to those of the parent and the current clinical aminoglycoside antibiotics gentamicin and tobramycin, as borne out by ex vivo studies with mouse cochlear explants. The 6'- N-(2-hydroxyethyl) modification restores antibacterial activity in the presence of the AAC(6') aminoglycoside-modifying enzymes, while the 1- N-(4-amino-2 S-hydroxybutyryl) modification overcomes resistance to the AAC(2') class but is still affected to some extent by the AAC(3) class. Neither modification is able to circumvent the ArmA ribosomal methyltransferase-induced aminoglycoside resistance. The use of phenyltriazenyl protection for the secondary amino group of sisomicin facilitates the synthesis of each derivative and their characterization through the provision of sharp NMR spectra for all intermediates.


Subject(s)
Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Ribosomes/physiology , Sisomicin/chemistry , Sisomicin/pharmacology , Aminoglycosides/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Base Sequence , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Escherichia coli/drug effects , Humans , Microbial Sensitivity Tests , Protein Biosynthesis/drug effects , Sisomicin/chemical synthesis , Structure-Activity Relationship
17.
Brain Res ; 1148: 83-9, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17359945

ABSTRACT

To define the role of free radical formation and potential energy depletion in noise induced hearing loss (NIHL), we measured the effectiveness of tempol (free radical scavenger) and creatine (enhances cellular energy storage) alone and in combination to attenuate NIHL. Guinea pigs were divided into four treatment groups: controls, 3% creatine diet (2 weeks prior to noise exposure), tempol (3 mM in drinking water 2 weeks prior to exposure), and creatine plus tempol and exposed to 120 dB SPL one-octave band noise centered at 4 kHz for 5 h. The noise-only control group showed frequency-dependent auditory threshold shifts (measured by auditory brainstem response, ABR) of up to 73 dB (16 kHz) on day 1, and up to 50 dB (8 kHz) on day 10. Creatine-treated subjects had significantly smaller ABR threshold shifts on day 1 and on day 10. Tempol alone significantly reduced ABR threshold shifts on day 10 but not on day 1. ABR shifts after combination treatment were similar to those in the creatine group. Hair cell loss on day 10 was equally attenuated by creatine and tempol alone or in combination. Our results indicate that the maintenance of ATP levels is important in attenuating both temporary and permanent NIHL, while the scavenging of free radicals provides protection from permanent NIHL.


Subject(s)
Cochlea/drug effects , Creatine/pharmacology , Cyclic N-Oxides/pharmacology , Free Radical Scavengers/pharmacology , Hearing Loss, Noise-Induced/drug therapy , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Auditory Threshold/drug effects , Auditory Threshold/physiology , Cochlea/metabolism , Cochlea/physiopathology , Creatine/therapeutic use , Cyclic N-Oxides/therapeutic use , Drug Synergism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/physiology , Free Radical Scavengers/therapeutic use , Free Radicals/antagonists & inhibitors , Free Radicals/metabolism , Guinea Pigs , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/physiopathology , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Noise/adverse effects , Oxidative Stress/physiology , Spin Labels , Treatment Outcome
18.
Hear Res ; 226(1-2): 178-82, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16844331

ABSTRACT

This article reviews recent advances in the protection from the adverse auditory or vestibular side effects associated with antibacterial treatment with aminoglycoside antibiotics. Compelling evidence from animal models suggests that reactive oxygen species are part of the initial mechanisms that trigger apoptotic and necrotic cell death in the inner ear. Consequently, antioxidants protect against aminoglycoside-induced hearing loss in animals and, importantly, they do so without compromising drug serum levels or antibacterial efficacy. While clinical studies have long confirmed the ototoxicity of aminoglycosides in human, a trial on protection was only recently reported (Sha, S.-H., Qiu, J.-H., Schacht, J., 2006. Aspirin attenuates gentamicin-induced hearing loss. New Engl. J. Med. 354, 1856-1857). Based on the finding that salicylate afforded protection in animals, the efficacy of aspirin (acetyl salicylate) was tested in a randomized double-blind placebo-controlled study in patients receiving gentamicin for acute infections. Fourteen of 106 patients (13%) met the criterion of hearing loss in the placebo group while only 3/89 (3%) were affected in the aspirin group (p=0.013). Aspirin did not influence gentamicin serum levels or the course of therapy. These results indicate that therapeutic protection from aminoglycoside ototoxicity may be extrapolated from animal models to the clinic. Furthermore, medications as common as aspirin can significantly attenuate the risk of gentamicin-induced hearing loss.


Subject(s)
Aspirin/pharmacology , Cochlea/drug effects , Gentamicins/antagonists & inhibitors , Gentamicins/toxicity , Vestibule, Labyrinth/drug effects , Animals , Anti-Bacterial Agents/antagonists & inhibitors , Anti-Bacterial Agents/toxicity , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Humans , Randomized Controlled Trials as Topic
19.
Expert Opin Investig Drugs ; 26(1): 85-96, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27918210

ABSTRACT

INTRODUCTION: Noise-induced hearing loss (NIHL) due to industrial, military, and recreational noise exposure is a major, but also potentially preventable cause of acquired hearing loss. For the United States it is estimated that 26 million people (15% of the population) between the ages of 20 and 69 have a high-frequency NIHL at a detriment to the quality of life of the affected individuals and great economic cost to society. Areas covered: This review outlines the pathology and pathophysiology of hearing loss as seen in humans and animal models. Results from molecular studies are presented that have provided the basis for therapeutic strategies successfully applied to animals. Several compounds emerging from these studies (mostly antioxidants) are now being tested in field trials. Expert opinion: Although no clinically applicable intervention has been approved yet, recent trials are encouraging. In order to maximize protective therapies, future work needs to apply stringent criteria for noise exposure and outcome parameters. Attention needs to be paid not only to permanent NIHL due to death of sensory cells but also to temporary effects that may show delayed consequences. Existing results combined with the search for efficacious new therapies should establish a viable treatment within a decade.


Subject(s)
Antioxidants/therapeutic use , Hearing Loss, Noise-Induced/prevention & control , Quality of Life , Adult , Aged , Animals , Antioxidants/pharmacology , Hearing Loss, Noise-Induced/epidemiology , Hearing Loss, Noise-Induced/physiopathology , Humans , Middle Aged , Time Factors , Young Adult
20.
Front Cell Neurosci ; 11: 315, 2017.
Article in English | MEDLINE | ID: mdl-29114207

ABSTRACT

Previous studies have reported that modification of histones alters aminoglycoside-induced hair cell death and hearing loss. In this study, we investigated three FDA-approved histone deacetylase (HDAC) inhibitors (vorinostat/SAHA, belinostat, and panobinostat) as protectants against aminoglycoside-induced ototoxicity in murine cochlear explants and in vivo in both guinea pigs and CBA/J mice. Individually, all three HDAC inhibitors reduced gentamicin (GM)-induced hair cell loss in a dose-dependent fashion in explants. In vivo, however, treatment with SAHA attenuated neither GM-induced hearing loss and hair cell loss in guinea pigs nor kanamycin (KM)-induced hearing loss and hair cell loss in mice under chronic models of ototoxicity. These findings suggest that treatment with the HDAC inhibitor SAHA attenuates aminoglycoside-induced ototoxicity in an acute model, but not in chronic models, cautioning that one cannot rely solely on in vitro experiments to test the efficacy of otoprotectant compounds.

SELECTION OF CITATIONS
SEARCH DETAIL