Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 97
1.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article En | MEDLINE | ID: mdl-37047074

Nonsense mutations trigger premature translation termination and often give rise to prevalent and rare genetic diseases. Consequently, the pharmacological suppression of an unscheduled stop codon represents an attractive treatment option and is of high clinical relevance. At the molecular level, the ability of the ribosome to continue translation past a stop codon is designated stop codon readthrough (SCR). SCR of disease-causing premature termination codons (PTCs) is minimal but small molecule interventions, such as treatment with aminoglycoside antibiotics, can enhance its frequency. In this review, we summarize the current understanding of translation termination (both at PTCs and at cognate stop codons) and highlight recently discovered pathways that influence its fidelity. We describe the mechanisms involved in the recognition and readthrough of PTCs and report on SCR-inducing compounds currently explored in preclinical research and clinical trials. We conclude by reviewing the ongoing attempts of personalized nonsense suppression therapy in different disease contexts, including the genetic skin condition epidermolysis bullosa.


Codon, Nonsense , Genetic Diseases, Inborn , Peptide Chain Elongation, Translational , Precision Medicine , Rare Diseases , Suppression, Genetic , Animals , Humans , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Codon, Nonsense/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/therapy , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Nephritis, Hereditary/genetics , Nephritis, Hereditary/therapy , Nonsense Mediated mRNA Decay , Peptide Chain Elongation, Translational/drug effects , Precision Medicine/methods , Precision Medicine/trends , Rare Diseases/genetics , Rare Diseases/therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/therapy , Suppression, Genetic/drug effects , Suppression, Genetic/genetics , Peptide Chain Termination, Translational/drug effects , Aminoglycosides/pharmacology
2.
Nat Commun ; 12(1): 4358, 2021 07 16.
Article En | MEDLINE | ID: mdl-34272367

Premature termination codons (PTCs) prevent translation of a full-length protein and trigger nonsense-mediated mRNA decay (NMD). Nonsense suppression (also termed readthrough) therapy restores protein function by selectively suppressing translation termination at PTCs. Poor efficacy of current readthrough agents prompted us to search for better compounds. An NMD-sensitive NanoLuc readthrough reporter was used to screen 771,345 compounds. Among the 180 compounds identified with readthrough activity, SRI-37240 and its more potent derivative SRI-41315, induce a prolonged pause at stop codons and suppress PTCs associated with cystic fibrosis in immortalized and primary human bronchial epithelial cells, restoring CFTR expression and function. SRI-41315 suppresses PTCs by reducing the abundance of the termination factor eRF1. SRI-41315 also potentiates aminoglycoside-mediated readthrough, leading to synergistic increases in CFTR activity. Combining readthrough agents that target distinct components of the translation machinery is a promising treatment strategy for diseases caused by PTCs.


Codon, Nonsense/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/drug effects , Nonsense Mediated mRNA Decay , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/metabolism , Aminoglycosides/metabolism , Codon, Nonsense/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Genes, Reporter , Gentamicins/pharmacology , HEK293 Cells , Humans , Microsomes, Liver/drug effects , Peptide Termination Factors/genetics , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Ribosomes/metabolism , Structure-Activity Relationship
3.
Nucleic Acids Res ; 49(13): 7665-7679, 2021 07 21.
Article En | MEDLINE | ID: mdl-34157102

Deciphering translation is of paramount importance for the understanding of many diseases, and antibiotics played a pivotal role in this endeavour. Blasticidin S (BlaS) targets translation by binding to the peptidyl transferase center of the large ribosomal subunit. Using biochemical, structural and cellular approaches, we show here that BlaS inhibits both translation elongation and termination in Mammalia. Bound to mammalian terminating ribosomes, BlaS distorts the 3'CCA tail of the P-site tRNA to a larger extent than previously reported for bacterial ribosomes, thus delaying both, peptide bond formation and peptidyl-tRNA hydrolysis. While BlaS does not inhibit stop codon recognition by the eukaryotic release factor 1 (eRF1), it interferes with eRF1's accommodation into the peptidyl transferase center and subsequent peptide release. In human cells, BlaS inhibits nonsense-mediated mRNA decay and, at subinhibitory concentrations, modulates translation dynamics at premature termination codons leading to enhanced protein production.


Peptide Chain Elongation, Translational/drug effects , Peptide Chain Termination, Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Cryoelectron Microscopy , HeLa Cells , Humans , Nonsense Mediated mRNA Decay/drug effects , Nucleosides/chemistry , Nucleosides/pharmacology , Peptide Termination Factors/metabolism , Peptides/metabolism , Protein Synthesis Inhibitors/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/drug effects , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article En | MEDLINE | ID: mdl-33674389

Apidaecin (Api), an unmodified 18-amino-acid-long proline-rich antibacterial peptide produced by bees, has been recently described as a specific inhibitor of translation termination. It invades the nascent peptide exit tunnel of the postrelease ribosome and traps the release factors preventing their recycling. Api binds in the exit tunnel in an extended conformation that matches the placement of a nascent polypeptide and establishes multiple contacts with ribosomal RNA (rRNA) and ribosomal proteins. Which of these interactions are critical for Api's activity is unknown. We addressed this problem by analyzing the activity of all possible single-amino-acid substitutions of the Api variants synthesized in the bacterial cell. By conditionally expressing the engineered api gene, we generated Api directly in the bacterial cytosol, thereby bypassing the need for importing the peptide from the medium. The endogenously expressed Api, as well as its N-terminally truncated mutants, retained the antibacterial properties and the mechanism of action of the native peptide. Taking advantage of the Api expression system and next-generation sequencing, we mapped in one experiment all the single-amino-acid substitutions that preserve or alleviate the on-target activity of the Api mutants. Analysis of the inactivating mutations made it possible to define the pharmacophore of Api involved in critical interactions with the ribosome, transfer RNA (tRNA), and release factors. We also identified the Api segment that tolerates a variety of amino acid substitutions; alterations in this segment could be used to improve the pharmacological properties of the antibacterial peptide.


Antimicrobial Cationic Peptides , Escherichia coli , Peptide Chain Termination, Translational/drug effects , Protein Synthesis Inhibitors , Amino Acid Substitution , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Bees , Escherichia coli/genetics , Escherichia coli/metabolism , Mutation, Missense , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism
5.
Elife ; 92020 10 08.
Article En | MEDLINE | ID: mdl-33031031

Biochemical studies suggested that the antimicrobial peptide apidaecin (Api) inhibits protein synthesis by binding in the nascent peptide exit tunnel and trapping the release factor associated with a terminating ribosome. The mode of Api action in bacterial cells had remained unknown. Here genome-wide analysis reveals that in bacteria, Api arrests translating ribosomes at stop codons and causes pronounced queuing of the trailing ribosomes. By sequestering the available release factors, Api promotes pervasive stop codon bypass, leading to the expression of proteins with C-terminal extensions. Api-mediated translation arrest leads to the futile activation of the ribosome rescue systems. Understanding the unique mechanism of Api action in living cells may facilitate the development of new medicines and research tools for genome exploration.


Antimicrobial Cationic Peptides/pharmacology , Codon, Terminator/metabolism , Escherichia coli/drug effects , Genome, Bacterial/drug effects , Peptide Chain Termination, Translational/drug effects , Ribosomes/metabolism , Codon, Terminator/drug effects , Escherichia coli/metabolism , Ribosomes/drug effects
6.
Drug Metab Dispos ; 48(4): 317-325, 2020 04.
Article En | MEDLINE | ID: mdl-31980502

Ataluren is a unique small molecule developed for the treatment of diseases caused by nonsense mutations, which result in premature termination of ribosomal translation and lack of full-length protein production. This study investigated the in vivo metabolism and disposition of ataluren in mice, rats, dogs, and humans. After single oral administration of [14C]ataluren, the overall recovery of radioactivity was ≥93.7%, with approximately 39%, 17%-21%, 12%, and 55% in the urine and 54%, 70%-72%, 80%, and 47% in the feces from intact mice, rats, dogs, and humans, respectively. In bile duct-cannulated (BDC) rats, approximately 10%, 7%, and 82% of the dose was recovered in the urine, feces, and bile, respectively, suggesting that biliary secretion was a major route for the elimination of ataluren in the rats. Ataluren was extensively metabolized after oral administration, and the metabolic profiles of ataluren were quantitatively similar across all species. Unchanged ataluren was the dominant radioactive component in plasma. Ataluren acyl glucuronide was the most prominent metabolite in plasma of all species and the dominant metabolite in BDC rat bile and human urine, whereas the oxadiazole cleavage products were the major or prominent metabolites in the feces of all species. Overall, the results indicate that phase I metabolism is negligible and that the pathway largely involves glucuronidation. No other circulatory conjugation metabolite was detected across investigated species. SIGNIFICANCE STATEMENT: Ataluren is a novel carboxylic acid-containing small molecule drug for treating nonsense mutation Duchenne muscular dystrophy. In vivo metabolism and disposition after a single dose of the drug were investigated in mice, rats, dogs, and humans. Phase I metabolism of ataluren was negligible, and the pathway largely involves glucuronidation. No other circulatory conjugation metabolite was detected across investigated species.


Muscular Dystrophy, Duchenne/drug therapy , Oxadiazoles/pharmacokinetics , Administration, Oral , Adolescent , Adult , Animals , Codon, Nonsense , Dogs , Female , Healthy Volunteers , Humans , Male , Metabolic Clearance Rate , Mice , Mice, Transgenic , Middle Aged , Muscular Dystrophy, Duchenne/genetics , Oxadiazoles/administration & dosage , Peptide Chain Termination, Translational/drug effects , Rats , Tissue Distribution , Young Adult
7.
Sci Rep ; 9(1): 15424, 2019 10 28.
Article En | MEDLINE | ID: mdl-31659219

The bacterial ribosome is an important drug target for antibiotics that can inhibit different stages of protein synthesis. Among the various classes of compounds that impair translation there are, however, no known small-molecule inhibitors that specifically target ribosomal release factors (RFs). The class I RFs are essential for correct termination of translation and they differ considerably between bacteria and eukaryotes, making them potential targets for inhibiting bacterial protein synthesis. We carried out virtual screening of a large compound library against 3D structures of free and ribosome-bound RFs in order to search for small molecules that could potentially inhibit termination by binding to the RFs. Here, we report identification of two such compounds which are found both to bind free RFs in solution and to inhibit peptide release on the ribosome, without affecting peptide bond formation.


Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Peptide Termination Factors/chemistry , Ribosomes/chemistry , Thermus thermophilus/chemistry , Anti-Bacterial Agents/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/antagonists & inhibitors , Peptide Termination Factors/metabolism , Ribosomes/metabolism , Thermus thermophilus/metabolism
8.
Nucleic Acid Ther ; 29(4): 175-186, 2019 08.
Article En | MEDLINE | ID: mdl-31070517

Efforts to develop treatments for diseases caused by nonsense mutations have focused on identification of small molecules that promote translational read-through of messenger RNAs (mRNAs) harboring nonsense stop codons to produce full-length proteins. However, to date, no small molecule read-through drug has received FDA approval, probably because of a lack of balance between efficacy and safety. Depletion of translation termination factors eukaryotic release factor (eRF) 1 and eRF3a in cells was shown to promote translational read-through of a luciferase reporter gene harboring a nonsense mutation. In this study, we identified antisense oligonucleotides (ASOs) targeting translation termination factors and determined if ASO-mediated depletion of these factors could be a potentially effective and safe therapeutic approach for diseases caused by nonsense mutations. We found that ASO-mediated reduction of either eRF1 or eRF3a to 30%-40% of normal levels in the mouse liver is well tolerated. Hemophilia mice that express a mutant allele of human coagulation factor IX (FIX) containing nonsense mutation R338X were treated with eRF1- or eRF3a-ASO. We found that although eRF1- or eRF3a-ASO alone only elicited a moderate read-through effect on hFIX-R338X mRNA, both worked in synergy with geneticin, a small molecule read-through drug, demonstrating significantly increased production of functional full-length hFIX protein to levels that would rescue disease phenotypes in these mice. Overall our results indicate that modulating the translation termination pathway in the liver by ASOs may provide a novel approach to improving the efficacy of small molecule read-through drugs to treat human genetic diseases caused by nonsense mutations.


Factor IX/genetics , Hemophilia A/therapy , Oligonucleotides, Antisense/genetics , Peptide Chain Termination, Translational/genetics , Animals , Codon, Nonsense/genetics , Disease Models, Animal , Gentamicins/pharmacology , Hemophilia A/genetics , Hemophilia A/pathology , Humans , Liver/drug effects , Liver/metabolism , Mice , Molecular Targeted Therapy , Oligonucleotides, Antisense/therapeutic use , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics
9.
PLoS One ; 14(5): e0216423, 2019.
Article En | MEDLINE | ID: mdl-31120902

Nonsense mutations constitute ~10% of TP53 mutations in cancer. They introduce a premature termination codon that gives rise to truncated p53 protein with impaired function. The aminoglycoside G418 can induce TP53 premature termination codon readthrough and thus increase cellular levels of full-length protein. Small molecule phthalimide derivatives that can enhance the readthrough activity of G418 have also been described. To determine whether readthrough enhancers exist among drugs that are already approved for use in humans, we tested seven antimalarial drugs for readthrough of the common R213X TP53 nonsense mutation in HDQ-P1 breast cancer cells. Mefloquine induced no TP53 readthrough activity as a single agent but it strongly potentiated readthrough by G418. The two enantiomers composing pharmaceutical mefloquine potentiated readthrough to similar levels in HDQ-P1 cells and also in SW900, NCI-H1688 and HCC1937 cancer cells with different TP53 nonsense mutations. Exposure to G418 and mefloquine increased p53 phosphorylation at Ser15 and P21 transcript levels following DNA damage, indicating p53 produced via readthrough was functional. Mefloquine does not appear to enhance readthrough via lysosomotropic effects as it did not significantly affect lysosomal pH, the cellular levels of G418 or its distribution in organellar or cytosolic fractions. The availability of a readthrough enhancer that is already approved for use in humans should facilitate study of the therapeutic potential of TP53 readthrough in preclinical cancer models.


Antimalarials/pharmacology , Codon, Nonsense , Codon, Terminator , Gentamicins/pharmacology , Mefloquine/pharmacology , Peptide Chain Termination, Translational , Tumor Suppressor Protein p53 , HCT116 Cells , Humans , Peptide Chain Termination, Translational/drug effects , Peptide Chain Termination, Translational/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
10.
Pediatr Nephrol ; 34(5): 873-881, 2019 05.
Article En | MEDLINE | ID: mdl-30413946

BACKGROUND: Cystinosis is an ultrarare disorder caused by mutations of the cystinosin (CTNS) gene, encoding a cystine-selective efflux channel in the lysosomes of all cells of the body. Oral therapy with cysteamine reduces intralysosomal cystine accumulation and slows organ deterioration but cannot reverse renal Fanconi syndrome nor prevent the eventual need for renal transplantation. A definitive therapeutic remains elusive. About 15% of cystinosis patients worldwide carry one or more nonsense mutations that halt translation of the CTNS protein. Aminoglycosides such as geneticin (G418) can bind to the mammalian ribosome, relax translational fidelity, and permit readthrough of premature termination codons to produce full-length protein. METHODS: To ascertain whether aminoglycosides permit readthrough of the most common CTNS nonsense mutation, W138X, we studied the effect of G418 on patient fibroblasts. RESULTS: G418 treatment induced translational readthrough of CTNSW138X constructs transfected into HEK293 cells and expression of full-length endogenous CTNS protein in homozygous W138X fibroblasts. CONCLUSIONS: Reduction in intracellular cystine indicates that the CTNS protein produced is functional as a cystine transporter. Interestingly, similar effects were seen even in W138X compound heterozygotes. These studies establish proof-of-principle for the potential of aminoglycosides to treat cystinosis and possibly other monogenic diseases caused by nonsense mutations.


Amino Acid Transport Systems, Neutral/genetics , Cystinosis/drug therapy , Fibroblasts/drug effects , Gentamicins/pharmacology , Peptide Chain Termination, Translational/drug effects , Codon, Nonsense , Cystine/metabolism , Cystinosis/genetics , Fibroblasts/metabolism , Genetic Vectors/genetics , Gentamicins/therapeutic use , HEK293 Cells , Humans , Peptide Chain Termination, Translational/genetics , Plasmids/genetics , RNA, Messenger/analysis , Recombinant Proteins/genetics , Transfection
11.
Biochem Biophys Res Commun ; 502(3): 422-428, 2018 07 20.
Article En | MEDLINE | ID: mdl-29857001

The ACTN3 gene encodes α-actinin-3 protein, which stabilizes the contractile apparatus at the Z-line in skeletal muscle cell fast fibers. A nonsense mutation of the arginine (R) at the codon for amino acid 577 of the ACTN3 gene generates a premature termination codon (PTC) and produces the R577X polymorphism in humans (X specifies translational termination). The ACTN3 577X genotype abolishes α-actinin-3 protein production due to targeted degradation of the mutant transcript by the cellular nonsense-mediated mRNA decay (NMD) system, which requires mRNA splicing. In humans, α-actinin-3 deficiency can decrease sprinting and power performance as well as skeletal muscle mass and strength. Here we investigated whether suppression of the in-frame PTC induced by treatment with the aminoglycosides gentamicin and G418 that promote termination codon readthrough could allow production of full-length α-actinin-3 protein from ACTN3 577X. We constructed expression plasmids encoding mature mRNA that lacks introns or pre-mRNA, which carries introns for the ACTN3 577X gene (X and Xpre, respectively) and transfected the constructs into HEK293 cells. Similar constructs for the ACTN3 577R gene were used as controls. HEK293 cells carrying the X gene, but not the Xpre gene, expressed exogenous truncated α-actinin-3 protein, indicating NMD-mediated suppression of exogenous Xpre expression. Cells treated with aminoglycosides produced exogenous full-length α-actinin-3 protein in X-transfected cells, but not in Xpre-transfected cells. The NMD inhibitor caffeine prevented suppression of Xpre expression and thereby induced production of full-length α-actinin-3 protein in the presence of aminoglycoside. Together these results indicate that the ACTN3 R577X polymorphism could be a novel target for readthrough therapy, which may affect athletic and muscle performance in humans.


Actinin/biosynthesis , Actinin/genetics , Codon, Nonsense , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Caffeine/pharmacology , Codon, Nonsense/drug effects , Gentamicins/pharmacology , HEK293 Cells , Humans , Muscle, Skeletal/metabolism , Peptide Chain Termination, Translational/drug effects , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
12.
J Mol Biol ; 430(5): 591-593, 2018 03 02.
Article En | MEDLINE | ID: mdl-29366636

Understanding the mechanisms of inhibitors of translation termination may inform development of new antibacterials and therapeutics for premature termination diseases. We report the crystal structure of the potent termination inhibitor blasticidin S bound to the ribosomal 70S•release factor 1 (RF1) termination complex. Blasticidin S shifts the catalytic domain 3 of RF1 and restructures the peptidyl transferase center. Universally conserved uridine 2585 in the peptidyl transferase center occludes the catalytic backbone of the GGQ motif of RF1, explaining the structural mechanism of inhibition. Rearrangement of domain 3 relative to the codon-recognition domain 2 provides insight into the dynamics of RF1 implicated in termination accuracy.


Anti-Bacterial Agents/pharmacology , Protein Biosynthesis/drug effects , Bacterial Proteins/metabolism , Catalytic Domain/drug effects , Codon, Terminator/metabolism , Models, Molecular , Nucleosides/antagonists & inhibitors , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/drug effects , Peptidyl Transferases/metabolism , Protein Conformation , Ribosomes/drug effects , Ribosomes/metabolism
13.
Nucleic Acids Res ; 44(7): 3264-75, 2016 Apr 20.
Article En | MEDLINE | ID: mdl-27001509

The antibiotic drug fusidic acid (FA) is commonly used in the clinic against gram-positive bacterial infections. FA targets ribosome-bound elongation factor G (EF-G), a translational GTPase that accelerates both messenger RNA (mRNA) translocation and ribosome recycling. How FA inhibits translocation was recently clarified, but FA inhibition of ribosome recycling by EF-G and ribosome recycling factor (RRF) has remained obscure. Here we use fast kinetics techniques to estimate mean times of ribosome splitting and the stoichiometry of GTP hydrolysis by EF-G at varying concentrations of FA, EF-G and RRF. These mean times together with previous data on uninhibited ribosome recycling were used to clarify the mechanism of FA inhibition of ribosome splitting. The biochemical data on FA inhibition of translocation and recycling were used to model the growth inhibitory effect of FA on bacterial populations. We conclude that FA inhibition of translocation provides the dominant cause of bacterial growth reduction, but that FA inhibition of ribosome recycling may contribute significantly to FA-induced expression of short regulatory open reading frames, like those involved in FA resistance.


Anti-Bacterial Agents/pharmacology , Fusidic Acid/pharmacology , Peptide Elongation Factor G/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Ribosomal Proteins/antagonists & inhibitors , Ribosomes/drug effects , Bacteria/drug effects , Bacteria/growth & development , Guanosine Triphosphate/metabolism , Peptide Chain Elongation, Translational/drug effects , Peptide Chain Termination, Translational/drug effects
14.
PLoS Genet ; 10(9): e1004616, 2014 Sep.
Article En | MEDLINE | ID: mdl-25233460

Release factors (RFs) govern the termination phase of protein synthesis. Human mitochondria harbor four different members of the class 1 RF family: RF1Lmt/mtRF1a, RF1mt, C12orf65 and ICT1. The homolog of the essential ICT1 factor is widely distributed in bacteria and organelles and has the peculiar feature in human mitochondria to be part of the ribosome as a ribosomal protein of the large subunit. The factor has been suggested to rescue stalled ribosomes in a codon-independent manner. The mechanism of action of this factor was obscure and is addressed here. Using a homologous mitochondria system of purified components, we demonstrate that the integrated ICT1 has no rescue activity. Rather, purified ICT1 binds stoichiometrically to mitochondrial ribosomes in addition to the integrated copy and functions as a general rescue factor, i.e. it releases the polypeptide from the peptidyl tRNA from ribosomes stalled at the end or in the middle of an mRNA or even from non-programmed ribosomes. The data suggest that the unusual termination at a sense codon (AGA/G) of the oxidative-phosphorylation enzymes CO1 and ND6 is also performed by ICT1 challenging a previous model, according to which RF1Lmt/mtRF1a is responsible for the translation termination at non-standard stop codons. We also demonstrate by mutational analyses that the unique insertion sequence present in the N-terminal domain of ICT1 is essential for peptide release rather than for ribosome binding. The function of RF1mt, another member of the class1 RFs in mammalian mitochondria, was also examined and is discussed.


Codon, Terminator , Mitochondria/genetics , Mitochondria/metabolism , Peptide Chain Termination, Translational , Proteins/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Animals , Codon , Humans , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Models, Biological , Molecular Sequence Data , Peptide Chain Termination, Translational/drug effects , Protein Binding , Protein Biosynthesis , Protein Interaction Domains and Motifs , Proteins/chemistry , Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins , Sequence Alignment , Swine
15.
Nucleic Acids Res ; 41(1): 264-76, 2013 Jan 07.
Article En | MEDLINE | ID: mdl-23087377

Ribosomes, after one round of translation, must be recycled so that the next round of translation can occur. Complete disassembly of post-termination ribosomal complex (PoTC) in yeast for the recycling consists of three reactions: release of tRNA, release of mRNA and splitting of ribosomes, catalyzed by eukaryotic elongation factor 3 (eEF3) and ATP. Here, we show that translocation inhibitors cycloheximide and lactimidomycin inhibited all three reactions. Cycloheximide is a non-competitive inhibitor of both eEF3 and ATP. The inhibition was observed regardless of the way PoTC was prepared with either release factors or puromycin. Paromomycin not only inhibited all three reactions but also re-associated yeast ribosomal subunits. On the other hand, sordarin or fusidic acid, when applied together with eEF2/GTP, specifically inhibited ribosome splitting without blocking of tRNA/mRNA release. From these inhibitor studies, we propose that, in accordance with eEF3's known function in elongation, the release of tRNA via exit site occurs first, then mRNA is released, followed by the splitting of ribosomes during the disassembly of post-termination complexes catalyzed by eEF3 and ATP.


Fungal Proteins/metabolism , Peptide Chain Termination, Translational/drug effects , Peptide Elongation Factors/metabolism , Protein Synthesis Inhibitors/pharmacology , Saccharomyces cerevisiae/genetics , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Cycloheximide/pharmacology , Fusidic Acid/pharmacology , Indenes/pharmacology , Macrolides/pharmacology , Paromomycin/pharmacology , Peptide Elongation Factors/antagonists & inhibitors , Peptide Termination Factors/metabolism , Piperidones/pharmacology , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Saccharomyces cerevisiae/drug effects
16.
Trends Mol Med ; 18(11): 679-88, 2012 Nov.
Article En | MEDLINE | ID: mdl-23083810

Ten percent of inherited diseases are caused by premature termination codon (PTC) mutations that lead to degradation of the mRNA template and to the production of a non-functional, truncated polypeptide. In addition, many acquired mutations in cancer introduce similar PTCs. In 1999, proof-of-concept for treating these disorders was obtained in a mouse model of muscular dystrophy, when administration of aminoglycosides restored protein translation by inducing the ribosome to bypass a PTC. Since, many studies have validated this approach, but despite the promise of PTC readthrough therapies, the mechanisms of translation termination remain to be precisely elucidated before even more progress can be made. Here, we review the molecular basis for PTC readthrough in eukaryotes and describe currently available compounds with significant therapeutic potential for treating genetic disorders and cancer.


Genetic Diseases, Inborn/drug therapy , Neoplasms/drug therapy , Aminoglycosides/adverse effects , Aminoglycosides/pharmacology , Aminoglycosides/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Codon, Nonsense , Genetic Diseases, Inborn/genetics , Humans , Models, Molecular , Neoplasms/genetics , Nonsense Mediated mRNA Decay/drug effects , Peptide Chain Termination, Translational/drug effects , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , RNA Stability
17.
PLoS Genet ; 8(3): e1002608, 2012.
Article En | MEDLINE | ID: mdl-22479203

The efficiency of translation termination depends on the nature of the stop codon and the surrounding nucleotides. Some molecules, such as aminoglycoside antibiotics (gentamicin), decrease termination efficiency and are currently being evaluated for diseases caused by premature termination codons. However, the readthrough response to treatment is highly variable and little is known about the rules governing readthrough level and response to aminoglycosides. In this study, we carried out in-depth statistical analysis on a very large set of nonsense mutations to decipher the elements of nucleotide context responsible for modulating readthrough levels and gentamicin response. We quantified readthrough for 66 sequences containing a stop codon, in the presence and absence of gentamicin, in cultured mammalian cells. We demonstrated that the efficiency of readthrough after treatment is determined by the complex interplay between the stop codon and a larger sequence context. There was a strong positive correlation between basal and induced readthrough levels, and a weak negative correlation between basal readthrough level and gentamicin response (i.e. the factor of increase from basal to induced readthrough levels). The identity of the stop codon did not affect the response to gentamicin treatment. In agreement with a previous report, we confirm that the presence of a cytosine in +4 position promotes higher basal and gentamicin-induced readthrough than other nucleotides. We highlight for the first time that the presence of a uracil residue immediately upstream from the stop codon is a major determinant of the response to gentamicin. Moreover, this effect was mediated by the nucleotide itself, rather than by the amino-acid or tRNA corresponding to the -1 codon. Finally, we point out that a uracil at this position associated with a cytosine at +4 results in an optimal gentamicin-induced readthrough, which is the therapeutically relevant variable.


Amino Acids/genetics , Codon, Nonsense , Gentamicins , Peptide Chain Termination, Translational , RNA, Transfer/genetics , Cells, Cultured , Codon, Nonsense/drug effects , Codon, Terminator/drug effects , Cytosine , Gentamicins/pharmacology , Humans , Peptide Chain Termination, Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Uracil
18.
Med Sci (Paris) ; 28(2): 193-9, 2012 Feb.
Article Fr | MEDLINE | ID: mdl-22377308

Ten percent of human hereditary diseases are linked to nonsense mutations (premature termination codon). These mutations lead to premature translation termination, trigger the synthesis of a truncated protein and possibly lead to mRNA degradation by the NMD pathway (nonsense mediated mRNA decay). For the past ten years, therapeutic strategies have emerged which attempt to use molecules that facilitate tRNA incorporation at premature stop codon (readthrough), thus allowing for the synthesis of a full length protein. Molecules currently used for this approach are mostly aminoglycoside antibiotics (gentamicin, amikacin…) that bind the decoding center of the ribosome. This therapeutic approach has been studied for various genetic diseases including Duchenne muscular dystrophy (DMD) and cystic fibrosis. The feasibility of this approach depends on induced readthrough level, mRNA quantity, re-expressed protein functionality and characteristics of each disease.


Codon, Nonsense/physiology , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Peptide Termination Factors/physiology , Alleles , Codon, Nonsense/genetics , Genetic Diseases, Inborn/genetics , Humans , Models, Biological , Models, Molecular , Mutagenesis, Site-Directed/methods , Peptide Chain Termination, Translational/drug effects , Peptide Chain Termination, Translational/genetics , Peptide Termination Factors/genetics , Substrate Specificity/genetics , Transcription, Genetic/genetics , Transcription, Genetic/physiology
19.
Wiley Interdiscip Rev RNA ; 2(6): 837-52, 2011.
Article En | MEDLINE | ID: mdl-21976286

Suppression therapy is a treatment strategy for genetic diseases caused by nonsense mutations. This therapeutic approach utilizes pharmacological agents that suppress translation termination at in-frame premature termination codons (PTCs) to restore translation of a full-length, functional polypeptide. The efficiency of various classes of compounds to suppress PTCs in mammalian cells is discussed along with the current limitations of this therapy. We also elaborate on approaches to improve the efficiency of suppression that include methods to enhance the effectiveness of current suppression drugs and the design or discovery of new, more effective suppression agents. Finally, we discuss the role of nonsense-mediated mRNA decay (NMD) in limiting the effectiveness of suppression therapy, and describe tactics that may allow the efficiency of NMD to be modulated in order to enhance suppression therapy.


Codon, Nonsense , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Aminoglycosides/toxicity , Animals , Codon, Nonsense/drug effects , Codon, Nonsense/genetics , Codon, Nonsense/metabolism , Drug Design , Genetic Diseases, Inborn/metabolism , Humans , Models, Biological , Models, Genetic , Nonsense Mediated mRNA Decay/drug effects , Peptide Chain Termination, Translational/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Suppression, Genetic
20.
EMBO J ; 30(8): 1485-96, 2011 Apr 20.
Article En | MEDLINE | ID: mdl-21399613

We have characterized a 17-residue peptide, MgtL, which is translated specifically in high Mg(2+) from an open reading frame (ORF) embedded in the Mg(2+) riboswitch domain, previously identified in the 5' leader region of Mg(2+) transporter gene mgtA in Salmonella. We demonstrate that mgtL translation is required to prematurely terminate mgtA transcription. Abrogation of mgtL translation by mutation of its start codon results in transcription of the mgtA-coding region in high Mg(2+), suggesting that ribosome stalling is not required for preventing premature transcription termination. Consistently, the Mg(2+) riboswitch responds to cytoplasmic Mg(2+), but not to proline or arginine, both repeatedly present in the MgtL sequence, to mediate mgtL translation-coupled regulation. RNA structural probing and nucleotide substitution analysis show that the riboswitch loop A region alters base pairing in response to Mg(2+), and favours stem-loop A1 in high Mg(2+), subsequently opening the ribosome-binding sequence for mgtL translation. Presumably, mgtL ORF directs translation to localize a ribosome in cis to act on downstream RNA in a manner similar to some upstream ORFs in prokaryotes and eukaryotes.


Magnesium/pharmacology , Peptide Chain Termination, Translational/drug effects , Protein Sorting Signals/genetics , Ribosomes/physiology , Riboswitch/physiology , Transcription, Genetic/drug effects , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Codon, Initiator , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Open Reading Frames , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
...