U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Madame Curie Bioscience Database [Internet]. Austin (TX): Landes Bioscience; 2000-2013.

Cover of Madame Curie Bioscience Database

Madame Curie Bioscience Database [Internet].

Show details

Therapies of Nonsense-Associated Diseases

, , and .

Author Information and Affiliations

A large number of diseases are caused by premature stop mutations that often lead to a complete loss of protein function and a severe reduction in mRNA levels due to nonsense-mediated mRNA decay (NMD). Two main approaches to develop treatments for diseases caused by premature stop mutations have been investigated. The first is to reduce the efficiency of translation termination through the use of pharmacological agents or by the expression of suppressor tRNAs. The second approach is to replace the premature stop mutation with wild-type sequence using gene repair techniques. Although each of these approaches have been demonstrated using in vitro studies or mouse models, currently only strategies using pharmacological agents to suppress stop mutations have reached preliminary clinical trials. The future of suppression therapy will require finding ways to increase the efficacy of current compounds to suppress premature stop mutations without side effects, or to design or discover safe new compounds that suppress premature stop mutations with increased efficiency. In addition, combined therapies that simultaneously suppress a premature stop mutation and inhibit NMD of the nonsense-containing mRNA may be the most effective way to increase the efficiency of suppression therapy.

Introduction

A large number of diseases including cystic fibrosis, Duchenne muscular dystrophy, β-thalassemia, and many types of cancers are caused by the presence of premature stop mutations in mRNAs. Premature stop mutations can arise as a result of mutations within germline or somatic DNA, inaccurate or inefficient pre-mRNA splicing, or improper RNA editing. According to the Human Gene Mutation Database, 12% of all mutations reported are singlepoint mutations that result in a premature stop codon.1 If mutations that alter the translational reading frame such as deletions, insertions, and splicing mutations are also considered, premature stop mutations may be responsible for as many as one-third of all inherited genetic disorders or cancers.2 Furthermore, the disease phenotypes caused by premature stop mutations are frequently more severe than those that result from missense mutations, since premature stop mutations often result in a complete loss of protein function.

Suppression of Premature Stop Mutations

One approach to treat diseases that result from in-frame premature stop mutations is to reduce the efficiency of translation termination so production of some full-length, functional protein is restored. Translation termination in eukaryotic cells occurs when one of the three stop codons, UAA (ochre), UAG (amber), or UGA (opal), enters the ribosomal A site. Stop codon recognition is not carried out by codon-anticodon interactions since no tRNA anticodons are complementary to any of the stop codons. Rather, stop codon recognition is mediated by a protein known as eukaryotic release factor 1 (eRF1). eRF1 recognizes each of the three stop codons in the ribosomal A site,3,4 and RNA cross-linking studies suggest that this interaction is direct.5 Upon recognition of a stop codon, eRF1 transmits a signal to the ribosomal peptidyl transferase center that leads to release of the nascent polypeptide from the peptidyl-tRNA located in the ribosomal P site.6 Another eukaryotic release factor, eRF3, is a GTPase that binds eRF17 and facilitates the efficiency and accuracy of stop codon recognition.8

Under normal conditions, translation termination is a very efficient process with an estimated error rate of approximately 0.1%.9-12 However, eRF1 and near-cognate aminoacyl-tRNAs (aminoacyl-tRNAs with an anticodon complementary to two of the three nucleotides of the stop codon) normally compete for A site binding. Under certain conditions, the rate at which near-cognate aminoacyl-tRNAs successfully compete with eRF1 at a stop codon can be increased, resulting in incorporation of an amino acid carried by a near-cognate aminoacyl-tRNA into the nascent polypeptide.13 This process is termed “termination suppression” or “readthrough”. In the case of a premature stop mutation, readthrough normally results in the continued elongation of the polypeptide chain in the correct reading frame and the production of full-length protein.

Obvious perturbations to the efficiency of translation termination include mutations in the translational machinery such as ribosomal proteins,14-19 ribosomal RNAs (rRNAs),20-23 termination factors,24-31 and aminoacyl-tRNAs.32-35 Interestingly, the identity of the stop codon itself also affects termination efficiency. Generally, termination is most efficient at UAA stop codons, followed by the UAG and UGA stop codons. In addition, the sequence context both upstream and downstream of the stop codon also influences termination efficiency.12,36-42 In particular, the first nucleotide downstream of the stop codon plays an important role in determining the efficiency of translation termination, and sequence analysis of natural stop codons has revealed a strong bias at that position in many species (including humans). This observation led to the proposal that eRF1 may normally recognize a tetranucleotide termination signal.42 RNA cross-linking studies have confirmed that eRF1 contacts the first nucleotide following the stop codon.43

Aminoglycoside-Mediated Nonsense Suppression

The efficiency of translation termination can also be reduced through the action of a large class of structurally related antibiotics called aminoglycosides. Aminoglycosides bind to a region of the small subunit rRNA known as the decoding site44 that normally monitors proper codon-anticodon interactions. Several nucleotides in the decoding site act to probe the conformation of the codon-anticodon helix to ensure that tRNA selection is correct. When aminoglycosides bind to the decoding site, they induce a conformational change that reduces the ability of rRNA to discriminate between cognate and near-cognate aminoacyl-tRNAs.45-50 This reduction in the accuracy of codon recognition increases the probability that translational misreading will occur, including the readthrough of stop codons.

Remarkably, the rRNA sequences that make up the prokaryotic and eukaryotic decoding sites are very similar (Fig. 1). One of the main differences lies within the major groove of the decoding site where aminoglycosides bind. A key residue for aminoglycoside binding to the prokaryotic decoding site, the A1408 nucleotide, is a G nucleotide in the corresponding position of the eukaryotic decoding site.51 Introduction of the A1408G mutation in the bacterial decoding site has been shown to reduce the affinity for aminoglycoside binding significantly.52 This suggests that this key nucleotide difference at least partially accounts for the specificity of aminoglycosides to inhibit the bacterial ribosome, resulting in their utility as antibiotics in humans.

Figure 1. Comparison of the decoding sites in E.

Figure 1

Comparison of the decoding sites in E. coli 16S rRNA and human 18S rRNA. Residues in the E. coli decoding site that are protected from dimethyl sulfoxide (DMS) modification by paromomycin are circled. Residues that are protected from DMS modification (more...)

However, several studies have shown that some aminoglycosides can also stimulate low levels of misreading that leads to termination suppression in eukaryotic systems.53-63 A recent yeast study revealed that aminoglycosides induced little or no misreading at sense codons, while the suppression of nonsense mutations was generally robust.64 This result suggests that stop codons are generally much more susceptible to aminoglycoside-induced misreading than sense codons in eukaryotes. This selectivity could be due to inherent differences in the fidelity of the elongation and termination processes.

The observations that aminoglycosides suppress premature stop codons in eukaryotic systems have led to a number of investigations to determine whether aminoglycosides could provide sufficient readthrough of premature stop mutations to suppress the phenotypes associated with human diseases. In most cases this question remains to be answered. However, it has been shown in mammalian cells that aminoglycosides can induce the suppression of nonsense mutations that cause many diseases, resulting in the restoration of low levels of functional protein. Aminoglycosides have been shown to induce readthrough of nonsense mutations that cause cystic fibrosis,65-67 Duchenne muscular dystrophy,68,69 Hurler syndrome,70,71 infantile neuronal ceroid lipofuscinosis,72 cystinosis,73 X-linked nephrogenic diabetes insipidus,74 recessive spinal muscular atrophy,75 and polycystic kidney disease.76 They have also been shown to suppress nonsense mutations in the p53 60 and ATM 77 tumor suppressor genes.

Particularly promising results have been obtained using mouse models for both Duchenne muscular dystrophy (DMD) and cystic fibrosis (CF). A mouse model for Duchenne muscular dystrophy known as the mdx mouse carries a naturally occurring UAA nonsense mutation in the dystrophin gene. It was shown that the administration of gentamicin via subcutaneous injections resulted in the partial restoration of dystrophin protein in muscle tissue of the mdx mouse.69 In addition, muscle contraction assays demonstrated that gentamicin treatment restored enough functional dystrophin protein to significantly reduce muscle contractile injury that is the hallmark of DMD. In another study, a transgenic CF mouse model that carried a UGA nonsense mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene was used to examine whether subcutaneous injections of gentamicin could restore CFTR protein expression. Treated mice showed a partial restoration of CFTR protein expression by immunofluoresence (Fig. 2) and a partial restoration of cAMP-activated chloride channel activity.78 These findings indicate that the administration of gentamicin stimulated readthrough of the CFTR premature stop mutation, resulting in the production of functional CFTR protein.

Figure 2. Immunofluorescence staining of submucosal glands in the duodenum shows the appearance of cystic fibrosis transmembrane conductance regulator (CFTR) protein following gentamicin treatment.

Figure 2

Immunofluorescence staining of submucosal glands in the duodenum shows the appearance of cystic fibrosis transmembrane conductance regulator (CFTR) protein following gentamicin treatment. Samples from homozygous Cftr-/- hCFTR-G542X transgenic mice (harvested (more...)

Several small clinical trials have also administered aminoglycosides to CF or DMD patients who carry premature stop mutations to determine whether any restoration of protein function occurred. To date, three such trials with CF patients have been reported. In two of the trials, the administration of gentamicin via nasal droplets restored some CFTR activity in the nasal epithelia of CF patients with a CFTR nonsense mutation.79,80 In a third study, a partial restoration of CFTR function was detected in the nasal epithelium of CF patients that carried a CFTR nonsense mutation when gentamicin was administered intravenously.81 Decidedly more mixed results were obtained in two clinical trials in which DMD patients with nonsense mutations were administered intravenous gentamicin. In one trial with four DMD patients, no increase in dystrophin levels or physical improvement could be ascertained after aminoglycoside treatment.82,83 However, the results of another clinical trial were more promising, since three of the four treated patients showed a partial restoration of dystrophin protein expression.83

The chemical structure of aminoglycosides determines their ability to suppress nonsense mutations (Fig. 3). Only a subset of these compounds is effective at suppressing nonsense mutations in eukaryotes, and only three aminoglycosides from this group (gentamicin, amikacin, and tobramycin) are approved for internal human use. In addition, the susceptibility of premature stop codons to aminoglycoside-mediated suppression depends on the identity of the stop codon and its surrounding mRNA sequence context.59,60 This codon and context dependence of aminoglycoside suppression is consistent with the results of a DMD clinical trial in which three DMD patients that carried UGA premature stop mutations showed a partial restoration of dystrophin expression after gentamicin treatment. However, no dystrophin could be detected in a patient with a UAA stop mutation after receiving the same gentamicin regimen.83 These results suggest that aminoglycoside suppression of a UGA stop codon may result in significantly more readthrough in patients than aminoglycoside suppression of a UAA codon, as previously concluded from in vitro studies.59,68,84 Thus, it may be necessary to alter the efficiency or stop codon specificity of nonsense suppression if a greater level of readthrough is to be obtained for diseases caused by certain stop mutations.

Figure 3. Structures of aminoglycosides commonly used in a clinical setting.

Figure 3

Structures of aminoglycosides commonly used in a clinical setting. The 2-deoxystreptamine ring is labeled ring II in each structure.

A major hurdle to the long-term use of aminoglycosides as a therapy to treat diseases caused by nonsense mutations is their toxicity, which can lead to kidney damage and hearing loss. However, the majority of these side effects do not appear to be due to their ability to induce translational misreading, but rather to other consequences related to their charged nature. Aminoglycosides are taken into cells via megalin, a multi-ligand, endocytic receptor that is particularly abundant in the proximal tubules of the kidney and the hair cells of the inner ear.85 Upon entering kidney cells, the positively charged nature of aminoglycoside molecules promotes their binding to acidic phospholipids in the lysosomal membrane,85,86 which alters the activity of a number of enzymes. In addition, aminoglycosides have been shown to promote the generation of free radical species that leads to tissue damage. Approaches that may reduce aminoglycoside toxicity include: altering the route and duration of their administration;87,88 coadministering compounds such as antioxidants to circumvent free radical damage;89-92 and coadministering polyanions such as poly-L-aspartate93,94 or daptomycin95,96 to sequester aminoglycosides away from the lysosomal membrane.

Another potential drawback associated with the suppression of premature stop mutations using aminoglycosides is the potential suppression of native stop codons. If this occurred on a global basis, it would lead to the production of many proteins with an extended C-terminus that could result in misfolding and loss of protein function. However, a previous study found that human cells cultured in the presence of high concentrations of aminoglycosides exhibited only a small increase in the level of the Hsp70 molecular chaperone, suggesting that little protein misfolding occurred during aminoglycoside treatment.70

The apparent lack of global readthrough at normal stop codons could be explained in several ways. Evidence of an evolutionary bias toward natural stop codons and surrounding sequence context(s) that may represent the most efficient termination signals has been observed at the end of genes in many species.42 No such selection for resistance to readthrough would occur at premature stop mutations. Thus, while the efficacy of aminoglycosides may be limited by the identity of the premature stop mutations and surrounding sequence context, these differences may also prevent readthrough at normal termination signals. In addition, multiple, in-frame stop codons are frequently found at the end of mRNAs.97-100 The presence of multiple stop codons should dramatically reduce the ability of aminoglycosides to induce readthrough of normal termination signals. Furthermore, the termination complex formed at premature stop codons appears to differ from the complex formed at native stop codons at the end of an mRNA.101,102 This intriguing finding suggests that the ribosome may terminate translation less efficiently at premature stop codons than native stop codons, possibly because the interactions between the termination complex at a premature stop codon and other factors bound in the 3' untranslated region of an mRNA cannot occur in a normal manner (see chapter by Amrani and Jacobson for further details).

Other Pharmacological Compounds That Suppress Nonsense Mutations

Since many of the toxic side effects caused by aminoglycosides are not directly associated with their ability to suppress stop mutations, another way to avoid these problems is to identify new, safer classes of compounds that suppress stop mutations. One such compound that has been investigated is negamycin. Negamycin is a dipeptide antibiotic that interacts with the ribosomal decoding site, much like aminoglycosides, even though it is structurally unrelated to aminoglycosides. Negamycin was shown to suppress the dystrophin premature stop mutation in the mdx mouse model, and was reported to be less toxic than aminoglycosides.103 Another drug, PTC124, is a novel compound discovered by PTC Therapeutics, Inc. that has been shown to suppress nonsense mutations in cell culture and in animal models.104 These results suggest that developing new pharmaceutical agents that suppress premature stop mutations without inducing the toxic side effects associated with aminoglycosides may have great potential for future therapeutic use.

Suppression of Nonsense Mutations Using Suppressor tRNAs

Another means of suppressing nonsense mutations involves expressing suppressor tRNAs. In this approach, DNA encoding a tRNA with an anticodon complementary to a stop codon is introduced into cells. This type of mutant tRNA, referred to as a suppressor tRNA, can compete with the termination factor eRF1 much more effectively than a near-cognate tRNA, resulting in a significant increase in stop codon suppression.

This approach has been shown to suppress premature stop mutations that cause β-thalassemia105 and Duchenne muscular dystrophy106 in mammalian cells. It has also been shown that the injection of DNA encoding a suppressor tRNA into the skeletal and heart muscles of a transgenic mouse expressing a reporter gene with a premature stop codon resulted in the suppression of the stop codon in vivo.107

Besides the potential suppression of natural stop codons that was discussed in a previous section, there are other significant drawbacks to this therapeutic approach. Although tRNA genes have strong RNA polymerase III promoters and the encoded tRNA molecules are generally stable after they are transcribed and undergo maturation,108 the efficient introduction and maintenance of tRNA genes into cells by gene therapy methods remains a challenge. In addition, the termination signal to be suppressed and its surrounding context also affect the efficiency of suppressor tRNA-mediated suppression.108,109

Mutation Repair

Another novel approach that could be used to treat diseases caused by premature stop mutations is the repair of a mutation directly in the genome using a nucleotide exchange antisense oligonucleotide approach. Unlike nonsense suppression therapy, which requires that the mutation be in the correct open reading frame, the use of antisense oligonucleotides also has the potential to repair various point mutations, small deletions, insertions, or splicing defects. In this method, a mutant DNA sequence is replaced by the wild-type sequence. Although a number of approaches have been designed to target DNA for mutation repair, including the use of ribozymes, group II introns, and triplex-forming oligonucleotides, single-stranded DNA currently appears to generate the most robust and reproducible gene repair. This exchange is directed by a double-stranded DNA chimeric oligonucleotide that is typically 70-80 nucleotides long. This chimera is synthesized as a single-stranded molecule that is designed with sequence complementarity such that is folds into a double-hairpin structure. The double-hairpin structure prevents the molecule from nuclease digestion as well as concatenation.110 When these molecules are introduced into cells, the chimeric oligonucleotide hybridizes to its complementary sequence in the target gene except for the region of mismatch sequence where the mutation lies. This mismatch is recognized by the cellular mismatch-repair system, which then catalyzes the exchange of the wild-type nucleotides for the mutant nucleotides, thus repairing the mutation in the DNA.

Gene repair of mutations has been demonstrated in yeast111,112 and mammalian cell culture, where correction of mutations that caused sickle cell anemia113 thalassemia,114 alkaline phosphatase deficiency,115 apolipoprotein A2-linked atherosclerosis,116 and epidermolysis bullosa simplex117 were accomplished. In addition, animal models of tyrosinemia,118 muscular dystrophy, 119-122 hemophilia,123 and renal tubular acidosis124 have been used to demonstrate a partial correction of mutations and a restoration of some protein expression. The amount of normal protein levels restored among the various disease phenotypes varied from 0.5% to 20% of wild-type levels.110 This level of functional protein could improve the phenotype of patients with these (and many other) genetic diseases.

Although this approach to correct mutations and restore wild-type protein production in mammalian cells is promising, its development for clinic application is still a work in progress. There are several problems that currently prevent the implementation of this approach.125 First, the design of stable oligonucleotides capable of replacing the mutation efficiently can be difficult. Second, as with many gene therapy approaches, the introduction of oligonucleotides into the appropriate cell type is a major obstacle, and the efficiency of repair is cell-cycle dependent. Third, the percentage of corrected cells decreases with time. Finally, apoptosis has been observed in some cells that have undergone this form of targeted sequence alteration. Thus, further studies are required to determine the ultimate utility of this approach.

Suppression of NMD

Many strategies aimed at suppressing premature stop mutations could be compromised by the fact that mRNAs that contain premature stop mutations are often unstable, resulting in a severe reduction in their steady-state level. The reduced abundance of mRNAs that carry a stop mutation is due to the NMD pathway (see chapter by Maquat). Therefore, approaches that stabilize nonsense-containing mRNAs that are normally degraded by NMD should increase the steady-state amount of mRNA available for translation. This, in turn, could greatly enhance the level of protein produced by suppression therapy.

The NMD pathway and the NMD factors Upf1, Upf2, and Upf3 are conserved in eukaryotes ranging from yeast to humans (see chapters by Baker and Parker, Singh and Lykke-Andersen, Anderson, and Behm-Ansmant and Izaurralde). Several factors involved in mammalian NMD bind to mRNAs in the nucleus during transcription and the subsequent stages of mRNA processing. In particular, some NMD components assemble with the exon-junction complex (EJC), located ˜20-24 nucleotides upstream of exon-exon junctions as a consequence of pre-mRNA splicing, and remain bound to the mRNA as it is exported to the cytoplasm126 (see chapter by Maquat). In human cells, Upf3 binds to nuclear mRNA-protein (mRNP) complexes as a component of the EJC complex. Upf2 binds to these complexes as they leave the nucleus, while Upf1 is thought to associate with the complex in the cytoplasm.127,128

According to current models, once the mRNA reaches the cytoplasm these bound nuclear factors are removed as the ribosome translates the mRNA during the initial or “pioneer round” of translation129,130 (see chapter by Maquat). If translation proceeds to the normal stop codon and all of the nuclear proteins are removed from the coding sequence during the pioneer round of translation, the transcript is remodeled to become steady-state mRNP and NMD can no longer occur. However, if a premature stop codon is present in the mRNA, the ribosome will not remove any nuclear proteins distal to the premature stop codon during this initial round of translation. This causes the mRNA to be identified as faulty and results in its rapid degradation by the NMD pathway. Generally, NMD occurs if an mRNA molecule carries a premature stop codon that is ≥50 nucleotides upstream of the 3'-most exon-exon junction.131

The destabilization of a nonsense-containing mRNA by NMD requires active translation of that mRNA. This conclusion is supported by the observations that several proteins that function in the NMD pathway associate with polysomes,132-135 and NMD can be inhibited by translation elongation inhibitors such as cycloheximide and puromycin. In yeast, the NMD factors have been shown to associate with the translation termination factors eRF1 and eRF3136 (see chapters by Baker and Parker, and Amrani and Jacobson). Of the Upf factors, at least Upf1 associates with eRF1 and eRF3 in mammals (see chapters by Maquat, and Singh and Lykke-Andersen), also indicating that NMD and the process of translation termination are likely to be tightly linked.

It has also been shown that suppression of a premature stop codon can stabilize the mRNA. Overexpression of a suppressor tRNA can inhibit the degradation of nonsense-containing mRNAs in yeast and in mammalian cells,137,138 and suppression of premature stop codons by aminoglycosides has also been shown to stabilize nonsense-containing mRNAs in some cases. For example, a 5-fold increase in the abundance of CFTR mRNA containing a premature stop codon was detected in a human bronchial epithelial cell line derived from a CF patient that carried the CFTR-W1282X premature stop mutation after incubation with the aminoglycoside G418 for 24 hours67 (Fig. 4). A 2-fold increase in mRNA was observed in human fibroblasts from a patient with Smith-Lemli-Opitz syndrome that carried a premature termination codon in the 7-dehydrocholesterol-delta 7-reductase (DHCR7) gene after G418 treatment.139 Finally, a 2-fold increase in mRNA was observed in human fibroblasts derived from a patient with Hurler syndrome that carried nonsense mutations in the α-L-iduronidase (IDUA) gene after gentamicin treatment (K.M. Keeling and D.M. Bedwell, unpublished data). These results are surprising since aminoglycosides only induced readthrough of these premature stop mutations at a low frequency of 1-20%. While it is possible that premature stop mutations are more susceptible to aminoglycoside-mediated suppression during the pioneer round of translation than in subsequent rounds of translation, the mechanism by which such a modest level of readthrough can inhibit NMD is currently unknown.

Figure 4. CFTR-W1282X mRNA abundance is increased in the IB3-1 bronchial epithelial cell line following G418 treatment.

Figure 4

CFTR-W1282X mRNA abundance is increased in the IB3-1 bronchial epithelial cell line following G418 treatment. IB3-1 cells carry the W1282X premature stop mutation on one CFTR allele and the ΔF508 mutation on the other CFTR allele. The ΔF508 (more...)

Certain factors in the NMD pathway have also been identified as potential targets for pharmacological inhibition of mRNA degradation by the NMD pathway. One such target is Upf1, which is essential for the NMD process and is conserved in all eukaryotes. Upf1 is a phosphoprotein, and changes in its phosphorylation state regulate its function in the NMD pathway.140,141 Several modifiers of the Upf1 phosphorylation state have been identified. For example, SMG1 is a kinase that phosphorylates at least two serine residues in Upf1 and stimulates the NMD process142-144 (see chapter by Yamashita et al). SMG1 is a member of the phosphatidylinositol-4,5-bisphosphate (PIP2) family of protein kinases, which are frequently inhibited by caffeine and wortmannin. Accordingly, these compounds were found to abrogate the degradation of the collagen VI α2 subunit mRNA by NMD in human fibroblasts, presumably by inhibiting SMG1 activity.145 The phosphorylation status of Upf1 is also influenced by SMG5, SMG6 and SMG7, which participate in the dephosphorylation of Upf1 and thus complete its phosphorylation-dephosphorylation cycle.146-149 These Upf1 modifiers also represent potential targets for pharmacological inhibition of NMD. Although the yeast Upf1 protein is also a phosphoprotein (K.M. Keeling and D.M. Bedwell, unpublished data), the kinase and phosphatase involved in its regulation have yet to be identified. Interestingly, a compound called diazoborine has been shown to stabilize aberrant mRNAs that are normally degraded by the NMD pathway in yeast.150 While the target of this compound is not yet known, it is possible that this compound could also target a component of the Upf1 phosphorylation cycle or some other step of yeast NMD.

It is important to note that the NMD pathway has additional functions other than degrading mRNAs that carry premature stop mutations (see chapters by Abraham and Oliveira, Azzalin et al, Kaygun and Marzluff, and Kim and Maquat). Mutations in Upf1, Upf2, and Upf3 influence the abundance of many normal mRNA species in yeast (see chapter by He and Jacobson) and mammals (see chapters by Sharifi and Dietz, and Soergel et al).151,152 Factors in the NMD pathway may also act as checkpoints for RNA processing and nuclear export.153,154 Therefore, therapeutic approaches aimed at inhibition of the NMD pathway must be monitored carefully and optimized such that other vital cellular pathways are not adversely affected.

Future Development of Therapies for Nonsense-Associated Diseases

Even though the results of several clinical trials indicate that the suppression of nonsense mutations can partially restore protein function, none of these studies were designed in a manner that allowed the investigators to determine whether enough protein expression was restored to confer a therapeutic improvement in the disease phenotype. The threshold level of functional protein needed to alleviate a disease phenotype is unknown for most diseases. This is a complex issue, since the minimal level of functional protein needed to improve a particular disease phenotype will vary widely depending upon the structure and function of each protein, and the key tissue(s) in which each protein is required. For example, estimates of the amount of CFTR protein required to alleviate the CF disease phenotype range from 5%155 to 30%156 of wild-type CFTR levels. In contrast, as little as 1% of α-L-iduronidase can reduce the severity of the Hurler syndrome disease phenotype.157,158 Therefore, suppression therapy may hold more potential for treating some disorders than others.

The development of new compounds or approaches that can suppress nonsense mutations without the side effects currently associated with aminoglycosides shows great potential for the future of nonsense suppression therapy. New pharmacological targets could include regions of rRNA, ribosomal proteins, translation termination factors, and components of the NMD machinery. Future therapies for the treatment of human diseases caused by premature stop mutations may include treatments designed to correct defects occurring at multiple levels simultaneously. For example, combining the suppression of premature stop mutations with the inhibition of NMD could restore a significantly higher level of functional protein than with either approach alone, thus providing a much greater opportunity to alleviate a disease phenotype. Currently, more research on the basic mechanisms of translation termination and NMD is needed to provide a better understanding of potential targets for therapies to treat nonsense-associated diseases.

References

1.
Krawczak M, Ball EV, Fenton I. et al. Human gene mutation database-a biomedical information and research resource. Hum Mutat. 2000;15(1):45–51. [PubMed: 10612821]
2.
Frischmeyer PA, Dietz HC. Nonsense-mediated mRNA decay in health and disease. Hum Mol Genet. 1999;8(10):1893–1900. [PubMed: 10469842]
3.
Kisselev L, Ehrenberg M, Frolova L. Termination of translation: Interplay of mRNA, rRNAs and release factors? EMBO J. 2003;22(2):175–182. [PMC free article: PMC140092] [PubMed: 12514123]
4.
Bertram G, Innes S, Minella O. et al. Endless possibilities: Translation termination and stop codon recognition. Microbiology. 2001;147(Pt 2):255–269. [PubMed: 11158343]
5.
Chavatte L, Frolova L, Kisselev L. et al. The polypeptide chain release factor eRF1 specifically contacts the s(4)UGA stop codon located in the A site of eukaryotic ribosomes. Eur J Biochem. 2001;268(10):2896–2904. [PubMed: 11358506]
6.
Frolova L, Le Goff X, Rasmussen HH. et al. A highly conserved eukaryotic protein family possessing properties of polypeptide chain release factor. Nature. 1994;372(6507):701–703. [PubMed: 7990965]
7.
Frolova L, Le Goff X, Zhouravleva G. et al. Eukaryotic polypeptide chain release factor eRF3 is an eRF1- and ribosome-dependent guanosine triphosphatase. RNA. 1996;2(4):334–341. [PMC free article: PMC1369376] [PubMed: 8634914]
8.
Salas-Marco J, Bedwell DM. GTP Hydrolysis by eRF3 facilitates stop codon decoding during eukaryotic translation termination. Mol Cell Biol. 2004;24(17):7769–7778. [PMC free article: PMC506980] [PubMed: 15314182]
9.
Stansfield I, Jones KM, Herbert P. et al. Missense translation errors in Saccharomyces cerevisiae. J Mol Biol. 1998;282(1):13–24. [PubMed: 9733638]
10.
Mori N, Funatsu Y, Hiruta K. et al. Analysis of translational fidelity of ribosomes with protamine messenger RNA as a template. Biochemistry. 1985;24(5):1231–1239. [PubMed: 4096904]
11.
Loftfield RB, Vanderjagt D. The frequency of errors in protein biosynthesis. Biochem J. 1972;128(5):1353–1356. [PMC free article: PMC1174024] [PubMed: 4643706]
12.
Bonetti B, Fu L, Moon J. et al. The efficiency of translation termination is determined by a synergistic interplay between upstream and downstream sequences in Saccharomyces cerevisiae. J Mol Biol. 1995;251(3):334–345. [PubMed: 7650736]
13.
Fearon K, McClendon V, Bonetti B. et al. Premature translation termination mutations are efficiently suppressed in a highly conserved region of yeast Ste6p, a member of the ATP-binding cassette (ABC) transporter family. J Biol Chem. 1994;269(27):17802–17808. [PubMed: 7517933]
14.
Zhang S, Ryden-Aulin M, Kirsebom LA. et al. Genetic implication for an interaction between release factor one and ribosomal protein L7/L12 in vivo. J Mol Biol. 1994;242(5):614–618. [PubMed: 7932718]
15.
Van Dyke N, Xu W, Murgola EJ. Limitation of ribosomal protein L11 availability in vivo affects translation termination. J Mol Biol. 2002;319(2):329–339. [PubMed: 12051910]
16.
Torres M, Condon C, Balada JM. et al. Ribosomal protein S4 is a transcription factor with properties remarkably similar to NusA, a protein involved in both nonribosomal and ribosomal RNA antitermination. EMBO J. 2001;20(14):3811–3820. [PMC free article: PMC125540] [PubMed: 11447122]
17.
Tate WP, Dognin MJ, Noah M. et al. The NH2-terminal domain of Escherichia coli ribosomal protein L11. Its three-dimensional location and its role in the binding of release factors 1 and 2. J Biol Chem. 1984;259(11):7317–7324. [PubMed: 6373771]
18.
Dahlgren A, Ryden-Aulin M. A novel mutation in ribosomal protein S4 that affects the function of a mutated RF1. Biochimie. 2000;82(8):683–691. [PubMed: 11018284]
19.
Brot N, Tate WP, Caskey CT. et al. The requirement for ribosomal proteins L7 and L12 in peptide-chain termination. Proc Natl Acad Sci USA. 1974;71(1):89–92. [PMC free article: PMC387938] [PubMed: 4589896]
20.
Velichutina IV, Hong JY, Mesecar AD. et al. Genetic interaction between yeast Saccharomyces cerevisiae release factors and the decoding region of 18 S rRNA. J Mol Biol. 2001;305(4):715–727. [PubMed: 11162087]
21.
Velichutina IV, Dresios J, Hong JY. et al. Mutations in helix 27 of the yeast Saccharomyces cerevisiae 18S rRNA affect the function of the decoding center of the ribosome. RNA. 2000;6(8):1174–1184. [PMC free article: PMC1369991] [PubMed: 10943896]
22.
Liu R, Liebman SW. A translational fidelity mutation in the universally conserved sarcin/ricin domain of 25S yeast ribosomal RNA. RNA. 1996;2(3):254–263. [PMC free article: PMC1369368] [PubMed: 8608449]
23.
Chernoff YO, Vincent A, Liebman SW. Mutations in eukaryotic 18S ribosomal RNA affect translational fidelity and resistance to aminoglycoside antibiotics. EMBO J. 1994;13(4):906–913. [PMC free article: PMC394890] [PubMed: 8112304]
24.
Zadorskii SP, Borkhsenius AS, Sopova Iu V. et al. Suppression of nonsense and frameshift mutations obtained by different methods for inactivating the translation termination factor eRF3 in yeast Saccharomyces cerevisiae. Genetika. 2003;39(4):489–494. [PubMed: 12760248]
25.
Wakem LP, Sherman F. Isolation and characterization of omnipotent suppressors in the yeast Saccharomyces cerevisiae. Genetics. 1990;124(3):515–522. [PMC free article: PMC1203945] [PubMed: 2179051]
26.
Vincent A, Newnam G, Liebman SW. The yeast translational allosuppressor, SAL6: A new member of the PP1-like phosphatase family with a long serine-rich N-terminal extension. Genetics. 1994;138(3):597–608. [PMC free article: PMC1206211] [PubMed: 7851758]
27.
Vincent A, Liebman SW. The yeast omnipotent suppressor SUP46 encodes a ribosomal protein which is a functional and structural homolog of the Escherichia coli S4 ram protein. Genetics. 1992;132(2):375–386. [PMC free article: PMC1205143] [PubMed: 1427034]
28.
Ono B, Tanaka M, Awano I. et al. Two new loci that give rise to dominant omnipotent suppressors in Saccharomyces cerevisiae. Curr Genet. 1989;16(5-6):323–330. [PubMed: 2692850]
29.
Liebman SW, Cavenagh M. An antisuppressor that acts on omnipotent suppressors in yeast. Genetics. 1980;95(1):49–61. [PMC free article: PMC1214221] [PubMed: 7000618]
30.
Kulikov VN, Tikhodeev ON, Forafonov FS. et al. Suppression of frameshift mutation as a result of partial inactivation of translation termination factors in Saccharomyces cerevisiae yeast. Genetika. 2001;37(5):602–609. [PubMed: 11436550]
31.
Carr-Schmid A, Valente L, Loik VI. et al. Mutations in elongation factor 1beta, a guanine nucleotide exchange factor, enhance translational fidelity. Mol Cell Biol. 1999;19(8):5257–5266. [PMC free article: PMC84369] [PubMed: 10409717]
32.
Valle RP, Morch MD, Haenni AL. Novel amber suppressor tRNAs of mammalian origin. EMBO J. 1987;6(10):3049–3055. [PMC free article: PMC553742] [PubMed: 3691479]
33.
Vacher J, Grosjean H, de Henau S. et al. Construction of a UGA suppressor tRNA by modification in vitro of yeast tRNACys. Eur J Biochem. 1984;138(1):77–81. [PubMed: 6363071]
34.
Kuchino Y, Beier H, Akita N. et al. Natural UAG suppressor glutamine tRNA is elevated in mouse cells infected with Moloney murine leukemia virus. Proc Natl Acad Sci USA. 1987;84(9):2668–2672. [PMC free article: PMC304719] [PubMed: 3472229]
35.
Beier H, Grimm M. Misreading of termination codons in eukaryotes by natural nonsense suppressor tRNAs. Nucleic Acids Res. 2001;29(23):4767–4782. [PMC free article: PMC96686] [PubMed: 11726686]
36.
Tork S, Hatin I, Rousset JP. et al. The major 5' determinant in stop codon read-through involves two adjacent adenines. Nucleic Acids Res. 2004;32(2):415–421. [PMC free article: PMC373328] [PubMed: 14736996]
37.
Namy O, Hatin I, Rousset JP. Impact of the six nucleotides downstream of the stop codon on translation termination. EMBO Rep. 2001;2(9):787–793. [PMC free article: PMC1084031] [PubMed: 11520858]
38.
Mottagui-Tabar S, Tuite MF, Isaksson LA. The influence of 5' codon context on translation termination in Saccharomyces cerevisiae. Eur J Biochem. 1998;257(1):249–254. [PubMed: 9799126]
39.
McCaughan KK, Brown CM, Dalphin ME. et al. Translational termination efficiency in mammals is influenced by the base following the stop codon. Proc Natl Acad Sci USA. 1995;92(12):5431–5435. [PMC free article: PMC41708] [PubMed: 7777525]
40.
Harrell L, Melcher U, Atkins JF. Predominance of six different hexanucleotide recoding signals 3' of read-through stop codons. Nucleic Acids Res. 2002;30(9):2011–2017. [PMC free article: PMC113845] [PubMed: 11972340]
41.
Cassan M, Rousset JP. UAG readthrough in mammalian cells: Effect of upstream and downstream stop codon contexts reveal different signals. BMC Mol Biol. 2001;2(1):3. [PMC free article: PMC29092] [PubMed: 11242562]
42.
Brown CM, Stockwell PA, Trotman CN. et al. Sequence analysis suggests that tetra-nucleotides signal the termination of protein synthesis in eukaryotes. Nucleic Acids Res. 1990;18(21):6339–6345. [PMC free article: PMC332501] [PubMed: 2123028]
43.
Bulygin KN, Repkova MN, Ven'yaminova AG. et al. Positioning of the mRNA stop signal with respect to polypeptide chain release factors and ribosomal proteins in 80S ribosomes. FEBS Lett. 2002;514(1):96–101. [PubMed: 11904189]
44.
Moazed D, Noller HF. Transfer RNA shields specific nucleotides in 16S ribosomal RNA from attack by chemical probes. Cell. 1986;47(6):985–994. [PubMed: 2430725]
45.
Yoshizawa S, Fourmy D, Puglisi JD. Structural origins of gentamicin antibiotic action. EMBO J. 1998;17(22):6437–6448. [PMC free article: PMC1170992] [PubMed: 9822590]
46.
Vicens Q, Westhof E. Crystal structure of paromomycin docked into the eubacterial ribosomal decoding A site. Structure (Camb). 2001;9(8):647–658. [PubMed: 11587639]
47.
Recht MI, Fourmy D, Blanchard SC. et al. RNA sequence determinants for aminoglycoside binding to an A-site rRNA model oligonucleotide. J Mol Biol. 1996;262(4):421–436. [PubMed: 8893854]
48.
Fourmy D, Yoshizawa S, Puglisi JD. Paromomycin binding induces a local conformational change in the A-site of 16 S rRNA. J Mol Biol. 1998;277(2):333–345. [PubMed: 9514734]
49.
Fourmy D, Recht MI, Puglisi JD. Binding of neomycin-class aminoglycoside antibiotics to the A-site of 16 S rRNA. J Mol Biol. 1998;277(2):347–362. [PubMed: 9514735]
50.
Fourmy D, Recht MI, Blanchard SC. et al. Structure of the A site of Escherichia coli 16S ribosomal RNA complexed with an aminoglycoside antibiotic. Science. 1996;274(5291):1367–1371. [PubMed: 8910275]
51.
Van de Peer Y, Van den Broeck I, De Rijk P. et al. Database on the structure of small ribosomal subunit RNA. Nucleic Acids Res. 1994;22(17):3488–3494. [PMC free article: PMC308309] [PubMed: 7524022]
52.
Recht MI, Douthwaite S, Puglisi JD. Basis for prokaryotic specificity of action of aminoglycoside antibiotics. EMBO J. 1999;18(11):3133–3138. [PMC free article: PMC1171394] [PubMed: 10357824]
53.
Wilhelm JM, Pettitt SE, Jessop JJ. Aminoglycoside antibiotics and eukaryotic protein synthesis: Structure-function relationships in the stimulation of misreading with a wheat embryo system. Biochemistry. 1978;17(7):1143–1149. [PubMed: 656378]
54.
Wilhelm JM, Jessop JJ, Pettitt SE. Aminoglycoside antibiotics and eukaryotic protein synthesis: Stimulation of errors in the translation of natural messengers in extracts of cultured human cells. Biochemistry. 1978;17(7):1149–1153. [PubMed: 656379]
55.
Stahl G, Bidou L, Rousset JP. et al. Versatile vectors to study recoding: Conservation of rules between yeast and mammalian cells. Nucleic Acids Res. 1995;23(9):1557–1560. [PMC free article: PMC306897] [PubMed: 7784210]
56.
Singh A, Ursic D, Davies J. Phenotypic suppression and misreading Saccharomyces cerevisiae. Nature. 1979;277(5692):146–148. [PubMed: 366438]
57.
Palmer E, Wilhelm JM, Sherman F. Phenotypic suppression of nonsense mutants in yeast by aminoglycoside antibiotics. Nature. 1979;277(5692):148–150. [PubMed: 366439]
58.
Palmer E, Wilhelm JM. Mistranslation in a eucaryotic organism. Cell. 1978;13(2):329–334. [PubMed: 75070]
59.
Manuvakhova M, Keeling K, Bedwell DM. Aminoglycoside antibiotics mediate context-dependent suppression of termination codons in a mammalian translation system. RNA. 2000;6(7):1044–1055. [PMC free article: PMC1369979] [PubMed: 10917599]
60.
Keeling KM, Bedwell DM. Clinically relevant aminoglycosides can suppress disease-associated premature stop mutations in the IDUA and p53 cDNAs in a mammalian translation system. J Mol Med. 2002;80(6):367–376. [PubMed: 12072912]
61.
Grentzmann G, Ingram JA, Kelly PJ. et al. A dual-luciferase reporter system for studying recoding signals. RNA. 1998;4(4):479–486. [PMC free article: PMC1369633] [PubMed: 9630253]
62.
Firoozan M, Grant CM, Duarte JA. et al. Quantitation of readthrough of termination codons in yeast using a novel gene fusion assay. Yeast. 1991;7(2):173–183. [PubMed: 1905859]
63.
Burke JF, Mogg AE. Suppression of a nonsense mutation in mammalian cells in vivo by the aminoglycoside antibiotics G-418 and paromomycin. Nucleic Acids Res. 1985;13(17):6265–6272. [PMC free article: PMC321951] [PubMed: 2995924]
64.
Salas-Marco J, Bedwell DM. Discrimination between defects in elongation fidelity and termination efficiency provides mechanistic insights into translational readthrough. J Mol Biol. 2005;348(4):801–815. [PubMed: 15843014]
65.
Zsembery A, Jessner W, Sitter G. et al. Correction of CFTR malfunction and stimulation of Ca-activated Cl channels restore HCO3- secretion in cystic fibrosis bile ductular cells. Hepatology. 2002;35(1):95–104. [PubMed: 11786964]
66.
Howard M, Frizzell RA, Bedwell DM. Aminoglycoside antibiotics restore CFTR function by overcoming premature stop mutations. Nat Med. 1996;2(4):467–469. [PubMed: 8597960]
67.
Bedwell DM, Kaenjak A, Benos DJ. et al. Suppression of a CFTR premature stop mutation in a bronchial epithelial cell line. Nat Med. 1997;3(11):1280–1284. [PubMed: 9359706]
68.
Howard MT, Shirts BH, Petros LM. et al. Sequence specificity of aminoglycoside-induced stop codon readthrough: Potential implications for treatment of Duchenne muscular dystrophy. Ann Neurol. 2000;48(2):164–169. [PubMed: 10939566]
69.
Barton-Davis ER, Cordier L, Shoturma DI. et al. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J Clin Invest. 1999;104(4):375–381. [PMC free article: PMC481050] [PubMed: 10449429]
70.
Keeling KM, Brooks DA, Hopwood JJ. et al. Gentamicin-mediated suppression of Hurler syndrome stop mutations restores a low level of alpha-L-iduronidase activity and reduces lysosomal glycosaminoglycan accumulation. Hum Mol Genet. 2001;10(3):291–299. [PubMed: 11159948]
71.
Hein LK, Bawden M, Muller VJ. et al. alpha-L-iduronidase premature stop codons and potential read-through in mucopolysaccharidosis type I patients. J Mol Biol. 2004;338(3):453–462. [PubMed: 15081804]
72.
Sleat DE, Sohar I, Gin RM. et al. Aminoglycoside-mediated suppression of nonsense mutations in late infantile neuronal ceroid lipofuscinosis. Eur J Paediatr Neurol. 2001;5(Suppl A):57–62. [PubMed: 11589009]
73.
Helip-Wooley A, Park MA, Lemons RM. et al. Expression of CTNS alleles: Subcellular localization and aminoglycoside correction in vitro. Mol Genet Metab. 2002;75(2):128–133. [PubMed: 11855931]
74.
Schulz A, Sangkuhl K, Lennert T. et al. Aminoglycoside pretreatment partially restores the function of truncated V(2) vasopressin receptors found in patients with nephrogenic diabetes insipidus. J Clin Endocrinol Metab. 2002;87(11):5247–5257. [PubMed: 12414899]
75.
Sossi V, Giuli A, Vitali T. et al. Premature termination mutations in exon 3 of the SMN1 gene are associated with exon skipping and a relatively mild SMA phenotype. Eur J Hum Genet. 2001;9(2):113–120. [PubMed: 11313744]
76.
Aguiari G, Banzi M, Gessi S. et al. Deficiency of polycystin-2 reduces Ca2+ channel activity and cell proliferation in ADPKD lymphoblastoid cells. FASEB J. 2004;18(7):884–886. [PubMed: 15001556]
77.
Lai CH, Chun HH, Nahas SA. et al. Correction of ATM gene function by aminoglycoside-induced read-through of premature termination codons. Proc Natl Acad Sci USA. 2004;101(44):15676–15681. [PMC free article: PMC524838] [PubMed: 15498871]
78.
Du M, Jones JR, Lanier J. et al. Aminoglycoside suppression of a premature stop mutation in a Cftr-/- mouse carrying a human CFTR-G542X transgene. J Mol Med. 2002;80(9):595–604. [PubMed: 12226741]
79.
Wilschanski M, Yahav Y, Yaacov Y. et al. Gentamicin-induced correction of CFTR function in patients with cystic fibrosis and CFTR stop mutations. N Engl J Med. 2003;349(15):1433–1441. [PubMed: 14534336]
80.
Wilschanski M, Famini C, Blau H. et al. A pilot study of the effect of gentamicin on nasal potential difference measurements in cystic fibrosis patients carrying stop mutations. Am J Respir Crit Care Med. 2000;161(3 Pt 1):860–865. [PubMed: 10712334]
81.
Clancy JP, Bebok Z, Ruiz F. et al. Evidence that systemic gentamicin suppresses premature stop mutations in patients with cystic fibrosis. Am J Respir Crit Care Med. 2001;163(7):1683–1692. [PubMed: 11401894]
82.
Wagner KR, Hamed S, Hadley DW. et al. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense mutations. Ann Neurol. 2001;49(6):706–711. [PubMed: 11409421]
83.
Politano L, Nigro G, Nigro V. et al. Gentamicin administration in Duchenne patients with premature stop codon. Preliminary results. Acta Myol. 2003;22(1):15–21. [PubMed: 12966700]
84.
Howard MT, Anderson CB, Fass U. et al. Readthrough of dystrophin stop codon mutations induced by aminoglycosides. Ann Neurol. 2004;55(3):422–426. [PubMed: 14991821]
85.
Nagai J, Takano M. Molecular aspects of renal handling of aminoglycosides and strategies for preventing the nephrotoxicity. Drug Metab Pharmacokinet. 2004;19(3):159–170. [PubMed: 15499183]
86.
Mingeot-Leclercq MP, Tulkens PM. Aminoglycosides: Nephrotoxicity. Antimicrob Agents Chemother. 1999;43(5):1003–1012. [PMC free article: PMC89104] [PubMed: 10223907]
87.
Beauchamp D, Labrecque G. Aminoglycoside nephrotoxicity: Do time and frequency of administration matter? Curr Opin Crit Care. 2001;7(6):401–408. [PubMed: 11805542]
88.
Bartal C, Danon A, Schlaeffer F. et al. Pharmacokinetic dosing of aminoglycosides: A controlled trial. Am J Med. 2003;114(3):194–198. [PubMed: 12637133]
89.
Sener G, Sehirli AO, Altunbas HZ. et al. Melatonin protects against gentamicin-induced nephrotoxicity in rats. J Pineal Res. 2002;32(4):231–236. [PubMed: 11982792]
90.
Nakashima T, Teranishi M, Hibi T. et al. Vestibular and cochlear toxicity of aminoglycosides - A review. Acta Otolaryngol. 2000;120(8):904–911. [PubMed: 11200584]
91.
Mazzon E, Britti D, De Sarro A. et al. Effect of N-acetylcysteine on gentamicin-mediated nephropathy in rats. Eur J Pharmacol. 2001;424(1):75–83. [PubMed: 11470263]
92.
Kawamoto K, Sha SH, Minoda R. et al. Antioxidant gene therapy can protect hearing and hair cells from ototoxicity. Mol Ther. 2004;9(2):173–181. [PubMed: 14759801]
93.
Gilbert DN, Wood CA, Kohlhepp SJ. et al. Polyaspartic acid prevents experimental aminoglycoside nephrotoxicity. J Infect Dis. 1989;159(5):945–953. [PubMed: 2651534]
94.
Beauchamp D, Laurent G, Maldague P. et al. Protection against gentamicin-induced early renal alterations (phospholipidosis and increased DNA synthesis) by coadministration of poly-L-aspartic acid. J Pharmacol Exp Ther. 1990;255(2):858–866. [PubMed: 2243354]
95.
Thibault N, Grenier L, Simard M. et al. Attenuation by daptomycin of gentamicin-induced experimental nephrotoxicity. Antimicrob Agents Chemother. 1994;38(5):1027–1035. [PMC free article: PMC188145] [PubMed: 8067733]
96.
Thibault N, Grenier L, Simard M. et al. Protection against gentamicin nephrotoxicity by daptomycin in nephrectomized rats. Life Sci. 1995;56(22):1877–1887. [PubMed: 7746096]
97.
Major LL, Edgar TD, Yee Yip P. et al. Tandem termination signals: Myth or reality? FEBS Lett. 2002;514(1):84–89. [PubMed: 11904187]
98.
Dalphin ME, Stockwell PA, Tate WP. et al. TransTerm, the translational signal database, extended to include full coding sequences and untranslated regions. Nucleic Acids Res. 1999;27(1):293–294. [PMC free article: PMC148161] [PubMed: 9847206]
99.
Dalphin ME, Brown CM, Stockwell PA. et al. The translational signal database, TransTerm, is now a relational database. Nucleic Acids Res. 1998;26(1):335–337. [PMC free article: PMC147192] [PubMed: 9399869]
100.
Brown CM, Dalphin ME, Stockwell PA. et al. The translational termination signal database. Nucleic Acids Res. 1993;21(13):3119–3123. [PMC free article: PMC309741] [PubMed: 8332534]
101.
Sachs MS, Wang Z, Gaba A. et al. Toeprint analysis of the positioning of translation apparatus components at initiation and termination codons of fungal mRNAs. Methods. 2002;26(2):105–114. [PubMed: 12054887]
102.
Amrani N, Ganesan R, Kervestin S. et al. A faux 3'-UTR promotes aberrant termination and triggers nonsense-mediated mRNA decay. Nature. 2004;432(7013):112–118. [PubMed: 15525991]
103.
Arakawa M, Shiozuka M, Nakayama Y. et al. Negamycin restores dystrophin expression in skeletal and cardiac muscles of mdx mice. J Biochem (Tokyo). 2003;134(5):751–758. [PubMed: 14688241]
104.
Hirawat S, Northcutt VJ, Welch EM. et al. Phase 1 safety and PK study of PTC124 for nonsense-mutation suppression therapy of cystic fibrosis. Pediatric Pulmonology. 2004;38(S27):248.
105.
Temple GF, Dozy AM, Roy KL. et al. Construction of a functional human suppressor tRNA gene: An approach to gene therapy for beta-thalassaemia. Nature. 1982;296(5857):537–540. [PubMed: 6803169]
106.
Kiselev AV, Ostapenko OV, Rogozhkina EV. et al. Suppression of nonsense mutations in the Dystrophin gene by a suppressor tRNA gene. Mol Biol (Mosk). 2002;36(1):43–47. [PubMed: 11862712]
107.
Buvoli M, Buvoli A, Leinwand LA. Suppression of nonsense mutations in cell culture and mice by multimerized suppressor tRNA genes. Mol Cell Biol. 2000;20(9):3116–3124. [PMC free article: PMC85606] [PubMed: 10757796]
108.
Atkinson J, Martin R. Mutations to nonsense codons in human genetic disease: Implications for gene therapy by nonsense suppressor tRNAs. Nucleic Acids Res. 1994;22(8):1327–1334. [PMC free article: PMC307985] [PubMed: 8190621]
109.
Phillips-Jones MK, Watson FJ. et al. The 3' codon context effect on UAG suppressor tRNA is different in Escherichia coli and human cells. J Mol Biol. 1993;233(1):1–6. [PubMed: 8377179]
110.
Kmiec EB. Targeted gene repair - In the arena. J Clin Invest. 2003;112(5):632–636. [PMC free article: PMC182220] [PubMed: 12952907]
111.
Rice MC, Czymmek K, Kmiec EB. The potential of nucleic acid repair in functional genomics. Nat Biotechnol. 2001;19(4):321–326. [PubMed: 11283588]
112.
Liu L, Rice MC, Kmiec EB. In vivo gene repair of point and frameshift mutations directed by chimeric RNA/DNA oligonucleotides and modified single-stranded oligonucleotides. Nucleic Acids Res. 2001;29(20):4238–4250. [PMC free article: PMC60207] [PubMed: 11600713]
113.
Cole-Strauss A, Yoon K, Xiang Y. et al. Correction of the mutation responsible for sickle cell anemia by an RNA-DNA oligonucleotide. Science. 1996;273(5280):1386–1389. [PubMed: 8703073]
114.
Dominski Z, Kole R. Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proc Natl Acad Sci USA. 1993;90(18):8673–8677. [PMC free article: PMC47420] [PubMed: 8378346]
115.
Kren BT, Cole-Strauss A, Kmiec EB. et al. Targeted nucleotide exchange in the alkaline phosphatase gene of HuH-7 cells mediated by a chimeric RNA/DNA oligonucleotide. Hepatology. 1997;25(6):1462–1468. [PubMed: 9185769]
116.
Tagalakis AD, Graham IR, Riddell DR. et al. Gene correction of the apolipoprotein (Apo) E2 phenotype to wild-type ApoE3 by in situ chimeraplasty. J Biol Chem. 2001;276(16):13226–13230. [PubMed: 11278248]
117.
D'Alessandro M, Morley SM, Ogden PH. et al. Functional improvement of mutant keratin cells on addition of desmin: An alternative approach to gene therapy for dominant diseases. Gene Ther. 2004;11(16):1290–1295. [PubMed: 15215887]
118.
Alexeev V, Yoon K. Stable and inheritable changes in genotype and phenotype of albino melanocytes induced by an RNA-DNA oligonucleotide. Nat Biotechnol. 1998;16(13):1343–1346. [PubMed: 9853616]
119.
Rando TA, Disatnik MH, Zhou LZ. Rescue of dystrophin expression in mdx mouse muscle by RNA/DNA oligonucleotides. Proc Natl Acad Sci USA. 2000;97(10):5363–5368. [PMC free article: PMC25834] [PubMed: 10805797]
120.
Mann CJ, Honeyman K, Cheng AJ. et al. Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proc Natl Acad Sci USA. 2001;98(1):42–47. [PMC free article: PMC14541] [PubMed: 11120883]
121.
Lu QL, Mann CJ, Lou F. et al. Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse. Nat Med. 2003;9(8):1009–1014. [PubMed: 12847521]
122.
Bertoni C, Rando TA. Dystrophin gene repair in mdx muscle precursor cells in vitro and in vivo mediated by RNA-DNA chimeric oligonucleotides. Hum Gene Ther. 2002;13(6):707–718. [PubMed: 11936970]
123.
Kren BT, Bandyopadhyay P, Steer CJ. In vivo site-directed mutagenesis of the factor IX gene by chimeric RNA/DNA oligonucleotides. Nat Med. 1998;4(3):285–290. [PubMed: 9500600]
124.
Lai LW, Chan DM, Erickson RP. et al. Correction of renal tubular acidosis in carbonic anhydrase II-deficient mice with gene therapy. J Clin Invest. 1998;101(7):1320–1325. [PMC free article: PMC508709] [PubMed: 9525974]
125.
Parekh-Olmedo H, Ferrara L, Brachman E. et al. Gene therapy progress and prospects: Targeted gene repair. Gene Ther. 2005;12(8):639–646. [PubMed: 15815682]
126.
Le Hir H, Moore MJ, Maquat LE. Pre-mRNA splicing alters mRNP composition: Evidence for stable association of proteins at exon-exon junctions. Genes Dev. 2000;14(9):1098–1108. [PMC free article: PMC316578] [PubMed: 10809668]
127.
Serin G, Gersappe A, Black JD. et al. Identification and characterization of human orthologues to Saccharomyces cerevisiae Upf2 protein and Upf3 protein (Caenorhabditis elegans SMG-4). Mol Cell Biol. 2001;21(1):209–223. [PMC free article: PMC88795] [PubMed: 11113196]
128.
Lykke-Andersen J, Shu MD, Steitz JA. Human Upf proteins target an mRNA for nonsense-mediated decay when bound downstream of a termination codon. Cell. 2000;103(7):1121–1131. [PubMed: 11163187]
129.
Lejeune F, Ishigaki Y, Li X. et al. The exon junction complex is detected on CBP80-bound but not eIF4E-bound mRNA in mammalian cells: Dynamics of mRNP remodeling. EMBO J. 2002;21(13):3536–3545. [PMC free article: PMC126094] [PubMed: 12093754]
130.
Ishigaki Y, Li X, Serin G. et al. Evidence for a pioneer round of mRNA translation: mRNAs subject to nonsense-mediated decay in mammalian cells are bound by CBP80 and CBP20. Cell. 2001;106(5):607–617. [PubMed: 11551508]
131.
Nagy E, Maquat LE. A rule for termination-codon position within intron-containing genes: When nonsense affects RNA abundance. Trends Biochem Sci. 1998;23(6):198–199. [PubMed: 9644970]
132.
Zhang J, Maquat LE. Evidence that translation reinitiation abrogates nonsense-mediated mRNA decay in mammalian cells. EMBO J. 1997;16(4):826–833. [PMC free article: PMC1169683] [PubMed: 9049311]
133.
Lim SK, Sigmund CD, Gross KW. et al. Nonsense codons in human beta-globin mRNA result in the production of mRNA degradation products. Mol Cell Biol. 1992;12(3):1149–1161. [PMC free article: PMC369546] [PubMed: 1545796]
134.
Atkin AL, Schenkman LR, Eastham M. et al. Relationship between yeast polyribosomes and Upf proteins required for nonsense mRNA decay. J Biol Chem. 1997;272(35):22163–22172. [PubMed: 9268361]
135.
Atkin AL, Altamura N, Leeds P. et al. The majority of yeast UPF1 colocalizes with polyribosomes in the cytoplasm. Mol Biol Cell. 1995;6(5):611–625. [PMC free article: PMC301219] [PubMed: 7545033]
136.
Czaplinski K, Ruiz-Echevarria MJ, Paushkin SV. et al. The surveillance complex interacts with the translation release factors to enhance termination and degrade aberrant mRNAs. Genes Dev. 1998;12(11):1665–1677. [PMC free article: PMC316864] [PubMed: 9620853]
137.
Gozalbo D, Hohmann S. Nonsense suppressors partially revert the decrease of the mRNA level of a nonsense mutant allele in yeast. Curr Genet. 1990;17(1):77–79. [PubMed: 2311129]
138.
Belgrader P, Cheng J, Maquat LE. Evidence to implicate translation by ribosomes in the mechanism by which nonsense codons reduce the nuclear level of human triosephosphate isomerase mRNA. Proc Natl Acad Sci USA. 1993;90(2):482–486. [PMC free article: PMC45687] [PubMed: 8421679]
139.
Correa-Cerro LS, Wassif CA, Waye JS. et al. DHCR7 nonsense mutations and characterisation of mRNA nonsense mediated decay in Smith-Lemli-Opitz syndrome. J Med Genet. 2005;42(4):350–357. [PMC free article: PMC1736027] [PubMed: 15805162]
140.
Page MF, Carr B, Anders KR. et al. SMG-2 is a phosphorylated protein required for mRNA surveillance in Caenorhabditis elegans and related to Upf1p of yeast. Mol Cell Biol. 1999;19(9):5943–5951. [PMC free article: PMC84455] [PubMed: 10454541]
141.
Bhattacharya A, Czaplinski K, Trifillis P. et al. Characterization of the biochemical properties of the human Upf1 gene product that is involved in nonsense-mediated mRNA decay. RNA. 2000;6(9):1226–1235. [PMC free article: PMC1369996] [PubMed: 10999600]
142.
Yamashita A, Ohnishi T, Kashima I. et al. Human SMG-1, a novel phosphatidylinositol 3-kinase-related protein kinase, associates with components of the mRNA surveillance complex and is involved in the regulation of nonsense-mediated mRNA decay. Genes Dev. 2001;15(17):2215–2228. [PMC free article: PMC312771] [PubMed: 11544179]
143.
Pal M, Ishigaki Y, Nagy E. et al. Evidence that phosphorylation of human Upfl protein varies with intracellular location and is mediated by a wortmannin-sensitive and rapamycin-sensitive PI 3-kinase-related kinase signaling pathway. RNA. 2001;7(1):5–15. [PMC free article: PMC1370068] [PubMed: 11214180]
144.
Grimson A, O'Connor S, Newman CL. et al. SMG-1 Is a Phosphatidylinositol kinase-related protein kinase required for nonsense-mediated mRNA decay in Caenorhabditis elegans. Mol Cell Biol. 2004;24(17):7483–7490. [PMC free article: PMC506987] [PubMed: 15314158]
145.
Usuki F, Yamashita A, Higuchi I. et al. Inhibition of nonsense-mediated mRNA decay rescues the phenotype in Ullrich's disease. Ann Neurol. 2004;55(5):740–744. [PubMed: 15122717]
146.
Ohnishi T, Yamashita A, Kashima I. et al. Phosphorylation of hUPF1 induces formation of mRNA surveillance complexes containing hSMG-5 and hSMG-7. Mol Cell. 2003;12(5):1187–1200. [PubMed: 14636577]
147.
Chiu SY, Serin G, Ohara O. et al. Characterization of human Smg5/7a: A protein with similarities to Caenorhabditis elegans SMG5 and SMG7 that functions in the dephosphorylation of Upf1. RNA. 2003;9(1):77–87. [PMC free article: PMC1370372] [PubMed: 12554878]
148.
Cali BM, Kuchma SL, Latham J. et al. smg-7 is required for mRNA surveillance in Caenorhabditis elegans. Genetics. 1999;151(2):605–616. [PMC free article: PMC1460488] [PubMed: 9927455]
149.
Anders KR, Grimson A, Anderson P. SMG-5, required for C. elegans nonsense-mediated mRNA decay, associates with SMG-2 and protein phosphatase 2A. EMBO J. 2003;22(3):641–650. [PMC free article: PMC140740] [PubMed: 12554664]
150.
Jungwirth H, Bergler H, Hogenauer G. Diazaborine treatment of Baker's yeast results in stabilization of aberrant mRNAs. J Biol Chem. 2001;276(39):36419–36424. [PubMed: 11477081]
151.
Lelivelt MJ, Culbertson MR. Yeast Upf proteins required for RNA surveillance affect global expression of the yeast transcriptome. Mol Cell Biol. 1999;19(10):6710–6719. [PMC free article: PMC84660] [PubMed: 10490610]
152.
He F, Li X, Spatrick P. et al. Genome-wide analysis of mRNAs regulated by the nonsense-mediated and 5' to 3' mRNA decay pathways in yeast. Mol Cell. 2003;12(6):1439–1452. [PubMed: 14690598]
153.
Reed R, Hurt E. A conserved mRNA export machinery coupled to pre-mRNA splicing. Cell. 2002;108(4):523–531. [PubMed: 11909523]
154.
Galy V, Gadal O, Fromont-Racine M. et al. Nuclear retention of unspliced mRNAs in yeast is mediated by perinuclear Mlp1. Cell. 2004;116(1):63–73. [PubMed: 14718167]
155.
Ramalho AS, Beck S, Meyer M. et al. Five percent of normal cystic fibrosis transmembrane conductance regulator mRNA ameliorates the severity of pulmonary disease in cystic fibrosis. Am J Respir Cell Mol Biol. 2002;27(5):619–627. [PubMed: 12397022]
156.
Kerem E. Pharmacologic therapy for stop mutations: How much CFTR activity is enough? Curr Opin Pulm Med. 2004;10(6):547–552. [PubMed: 15510065]
157.
Ashton LJ, Brooks DA, McCourt PA. et al. Immunoquantification and enzyme kinetics of alpha-L-iduronidase in cultured fibroblasts from normal controls and mucopolysaccharidosis type I patients. Am J Hum Genet. 1992;50(4):787–794. [PMC free article: PMC1682646] [PubMed: 1550122]
158.
Aronovich EL, Pan D, Whitley CB. Molecular genetic defect underlying alpha-L-iduronidase pseudodeficiency. Am J Hum Genet. 1996;58(1):75–85. [PMC free article: PMC1914939] [PubMed: 8554071]
159.
Purohit P, Stern S. Interactions of a small RNA with antibiotic and RNA ligands of the 30S subunit. Nature. 1994;370(6491):659–662. [PubMed: 8065453]
160.
Trapnell BC, Chu CS, Paakko PK. et al. Expression of the cystic fibrosis transmembrane conductance regulator gene in the respiratory tract of normal individuals and individuals with cystic fibrosis. Proc Natl Acad Sci USA. 1991;88(15):6565–6569. [PMC free article: PMC52127] [PubMed: 1713683]
161.
Hamosh A, Trapnell BC, Zeitlin PL. et al. Severe deficiency of cystic fibrosis transmembrane conductance regulator messenger RNA carrying nonsense mutations R553X and W1316X in respiratory epithelial cells of patients with cystic fibrosis. J Clin Invest. 1991;88(6):1880–1885. [PMC free article: PMC295756] [PubMed: 1721624]
162.
Hamosh A, Rosenstein BJ, Cutting GR. CFTR nonsense mutations G542X and W1282X associated with severe reduction of CFTR mRNA in nasal epithelial cells. Hum Mol Genet. 1992;1(7):542–544. [PubMed: 1284888]
Copyright © 2000-2013, Landes Bioscience.
Bookshelf ID: NBK6183

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...