You are on page 1of 5

nature publishing group DISCOVERY

Aminoglycoside-induced Translational
Read-through in Disease: Overcoming
Nonsense Mutations by Pharmacogenetic
Therapy
LV Zingman1,2, S Park1,2, TM Olson1,2, AE Alekseev1,2 and A Terzic1,2

A third of inherited diseases result from premature tumor-suppressor genes are also common in the develop-
termination codon mutations. Aminoglycosides have ment and progression of cancer.2
emerged as vanguard pharmacogenetic agents in treating At the molecular level, PTC mutations have major
human genetic disorders due to their unique ability to consequences. Typically, a PTC mutation precipitates rapid
suppress gene translation termination induced by nonsense degradation of the mRNA carrying the premature codon, after
mutations. In preclinical and pilot clinical studies, this a single cycle of translation, through nonsense-mediated
therapeutic approach shows promise in phenotype correction mRNA decay. Activation of this mechanism, while protective
by promoting otherwise defective protein synthesis. The against potentially lethal production of truncated protein by
challenge ahead is to maximize efficacy while preventing nullifying deleterious suppression of synthesis and/or function
interaction with normal protein production and function. of vital cell proteins, may not prevent the development of a
disease phenotype due to the absence of the translationally
defective protein itself.2,4,7 In the absence of nonsense-
The information encoded in DNA and the proper decoding mediated mRNA decay, truncated polypeptides could be
into proteins are fundamental to life.1 Change in the genetic produced.2,4,7 Assembly of a truncated protein in vivo is,
code brings the danger of life-threatening disease. There are however, a rare event, and is usually related to a PTC
more than 1,800 inherited human diseases caused by positioned within the last exon or in a splicing region resulting
nonsense mutations, i.e., alterations in the genetic code that in mutation-induced exon skipping. The biological effect of a
prematurely stop the translation of proteins.2,3 Indeed, about mutated gene may be difficult to predict, as it depends on the
30 percent of heritable disorders result from premature extent of protein truncation, stability of the polypeptide
termination codon (PTC) mutations.2,3 This frequency may product, and degree of interference with the expression of the
be even higher in certain syndromes or in specific human normal allele (e.g., dominant-negative effect).2,7
populations. Moreover, different classes of mutations, Two major therapeutic approaches to overcome nonsense
including frameshift insertions and/or deletions, splice-site mutations are presently considered. Gene therapy is, in
intron inclusions, or simple substitutions of a normal codon principle, the treatment of choice due to the potential for
with UAA, UAG, or UGA codons, can all introduce a targeted repair, but, to date, several limitations have
premature stop signal, thus arresting protein synthesis.4 Well- precluded clinical success.3,7 Alternatively, pharmacological
known examples include Duchenne muscular dystrophy, the approaches can modify gene expression and block nonsense-
most common X-linked fatal genetic disease, where 410–20 mediated mRNA destruction, without correcting the under-
percent of patients carry a nonsense mutation resulting in lying gene defect. The rationale supporting such emerging
premature translation termination. Similarly, in cystic pharmacogenetic strategies is the premise that even limited
fibrosis, B2–5 percent of patients (60 percent for Ashkenazi expression of a mutated gene could result in the production
Jews) have nonsense mutations in the cystic fibrosis of a partially or fully functional protein, sufficient for
transmembrane regulator gene.2,4–6 PTC mutations in therapeutic benefit.2,4,6,7

1
Marriott Heart Disease Research Program, Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA; 2Department
of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA. Correspondence: LV Zingman (zingman.leonid@mayo.edu)
doi:10.1038/sj.clpt.6100012

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 81 NUMBER 1 | JANUARY 2007 99


DISCOVERY

AMINOGLYCOSIDES: PROTEIN TRANSLATION-MODIFYING AMINOGLYCOSIDES: PHARMACOGENETIC AGENTS


DRUGS In 1985, Burke and Mogg12 were the first to demonstrate that
Aminoglycosides are widely used in clinical practice as the aminoglycoside antibiotics paromomycin and G-418
bactericidal antibiotics with established effects on transla- could partially restore the synthesis of a full-size protein from
tional accuracy or efficiency.8 Their utility as antibacterial a mutant gene with a premature UAG mutation in cultured
agents arises from binding to the highly conserved 16S mammalian cells. Later, G-418 and gentamicin were shown to
ribosomal RNA (rRNA) at the decoding center (Figure 1). restore the expression of the cystic fibrosis transmembrane
This center normally facilitates accurate codon–anticodon conductance regulator protein in a cell line carrying a
pairing. In the presence of an aminoglycoside, the conforma- nonsense mutation in cystic fibrosis transmembrane conduc-
tion of rRNA becomes altered, inducing codon misreading tance regulator.13,14 In 1999, the ability of aminoglycosides to
that causes either incorporation of an erroneous amino acid promote protein translation, despite the presence of a PTC
(mis-incorporation) at a sense codon or failure of recognition mutation, was first demonstrated in vivo in a model of
of the stop codon, leading to translational read-through Duchenne muscular dystrophy.15 In this way, the concept of
rather than chain termination (Figure 2).9–11 Although aminoglycoside-induced read-through emerged as a thera-
aminoglycosides target a conserved region of the rRNA peutic strategy in human genetic disorders. In the current era,
sequence, these agents are highly active against bacterial and aminoglycosides have been shown to suppress premature
mitochondrial ribosomes, but have limited interaction with translation termination at nonsense codons when bound to
human ribosomes. The specificity is related to the high- the rRNA decoding site interacting with codon–anticodon
affinity binding of aminoglycosides to prokaryotic rRNA, pairing in a series of clinically relevant conditions (Table 1).
which has an adenine at position 1408 of the 16S rRNA The efficiency of read-through varies from 1 percent to 25
(numbered according to the Escherichia coli sequence). In percent in human cell lines, largely depending on the context
contrast, eucaryotic ribosomes have a guanine at the of the stop mutation, with UGA showing greater translational
corresponding position, causing a low affinity towards read-through than UAG, and UAA exhibiting the most
aminoglycosides (Figure 2).11 The effects of aminoglycosides resistance to suppression.16,17 Several studies demonstrate the
on protein translation in eucaryotic cells have been demon- critical influence of upstream and downstream sequences on
strated at concentrations 10 to 15 times higher than the overall efficiency of translational termination.18,19 In mam-
typical therapeutic antibacterial concentrations. Partial sus- malian cells, the action of the antibiotic is governed by
ceptibility of eucaryotic ribosomes toward aminoglycoside nucleotides surrounding the stop mutation. The nucleotide
interaction has been traditionally viewed as the underlying in the position immediately downstream from the stop
mechanism of drug toxicity. However, this side effect may codon has a particularly significant impact on the efficiency
provide the opportunity for the treatment of human genetic of aminoglycoside-induced read-through, as follows:
diseases associated with translational defects. C4U4AXG.17

AMINOGLYCOSIDES: TRANSLATION OF PHARMACOGENETIC


AGENTS IN MEDICINE
Production of protein in the presence of a nonsense PTC
mutation resulting from aminoglycoside therapy, even if only
in low amount, may be functionally significant. This is
especially the case in recessive disorders, where protein
expression is essentially absent. In such cases, even 1 percent
of normal protein function may restore a near-normal or
clinically less severe phenotype.2,4,6,7 Accordingly, it is
primarily in recessive disorders that aminoglycosides have
provided the greatest promise in both cell culture experi-
ments and clinical trials. Examples include interventions with
defective genes in cystic fibrosis, Duchenne muscular
dystrophy, cystinosis, mucopolysaccharidosis type I (Hurler
syndrome), X-linked nephrogenic diabetes insipidus, ataxia
–telangiectasia, hemophilia, factor VII deficiency, and in-
fantile neuronal ceroid lipofuscinosis (Table 1).20–29
Recent studies have indicated that read-through induction
may be a promising therapy in autosomal-dominant
Figure 1 Structure of aminoglycoside bound to rRNA. Paromomycin (red) disorders as well (Table 1).30–32 This is somewhat surprising
binds to helix 44 in 16S rRNA. Adenine in position 1408 is critical for
high-affinity interaction with bacterial ribosome, with other residues
since haploinsufficiency is the common pathogenic mechan-
critical for codon–anticodon cognition also indicated. Atomic coordinates ism in nonsense mutation-related dominant disorders, i.e.,
are from 1IBK. B50 percent decrease in the amount of protein produced

100 VOLUME 81 NUMBER 1 | JANUARY 2007 | www.nature.com/cpt


DISCOVERY

Figure 2 Aminoglycoside interaction with ribosomal protein translation. (a–c) Interaction of paromomycin with 16S rRNA of the 30S ribosome subunit.
a In the absence of antibiotic, the conserved 16S rRNA (yellow strands) nucleotides A1492 and A1493 are stacked in the interior of helix 44. b Binding
of mRNA codon and cognate tRNA causes conformational change of A1492, A1493, and G530 to accommodate energetically favorable interactions of
ribosome components, tRNA and mRNA. c Paromomycin binding to the interior of helix44 induces a local conformational change in nucleotides A1492
and A1493, flipping them out. These changes facilitate binding of near-cognate tRNA. Black lines represent H-bonds. Crystal atomic coordinates are
from 1J5E, IBK, and 1IBM. (d–f) Aminoglycoside induced read-through of the premature E375X stop codon in Kv1.5. d Normally, tRNA carrying glutamic
acid (E) matches the mRNA codon to process Kv1.5 polypeptide elongation. Matching of the mRNA codon to the proper tRNA anticodon results in
conformational alignment of A1492 and A1493 in the ribosomal decoding center (red dashes) and polypeptide chain elongation. e E375X (UAA codon in
mRNA) mutation prevents codon–anticodon pairing and excludes the possibility of the A1492 and A1493 alignment in the ribosomal decoding center
(red dashes) terminating protein translation. f Aminoglycoside binding to 16S rRNA induces conformational alignment in the ribosomal decoding center
despite codon/anticodon mismatch. In the presence of aminoglycosides, the UAA codon may be paired with CUU or GUU tRNA anticodon promoting
polypeptide chain elongation with glutamate or glutamine.

due to the heterozygous loss of one allele causes clinical eluted patch atrial implantation could be considered for
symptoms. Just as a mutated protein can have a dominant- patients with PTC mutations causing atrial arrhythmias.
negative effect on the protein produced by the normal allele, Furthermore, after treatment with aminoglycosides, en-
even a small increase in full-length functional protein hanced production of functional protein from defective
resulting from aminoglycoside therapy may improve out- genes can be boosted through simultaneous application of
come.30–32 A case in point is Hailey–Hailey disease, caused by protein-specific promoters.35 Also, recent discoveries in the
a nonsense mutation in the Ca2 þ -ATPase2C1 transporter structure of ribosomes and their interactions with aminogly-
gene (ATP2C1),32 and familial atrial fibrillation related to a cosides provide a platform on which to engineer new
PTC mutation in KCNA5 encoding the voltage-sensitive generations of pharmacogenetic agents with tissue predilec-
potassium channel Kv1.5.31 In both cases, aminoglycosides tion and molecular specificity, and limit or prevent undesired
have been suggested as corrective therapy.30,31 interactions with normal protein synthesis and function.9–11
A limitation to aminoglycoside use as pharmacogenetic
agents is necessary to reach concentrations sufficient to CONCLUSION
interfere with protein synthesis at human ribosomes, Currently, aminoglycosides are the only clinically available
increasing risk of significant toxicity. Therefore, clinical drug family known to consistently affect gene translation in
success to date has been observed with local aminoglycoside eucaryotes. This property has provided the foundation for
delivery, particularly treatment of cystic fibrosis, the most preclinical and clinical testing of aminoglycosides to rescue
studied disease in the context of pharmacogenetic-based protein production arrested by nonsense mutations. Ami-
therapy. Systemic delivery of currently available aminoglyco- noglycosides have shown initial promise in diverse genetic
sides at clinically approved doses has failed, as in the case of disorders due to PTC mutations, particularly when applied
McArdle disease and Duchenne muscular dystrophy.33,34 locally to secure sufficient therapeutic levels while avoiding
Thus, developing effective drug delivery strategies that would systemic toxicity. The challenge in the emerging field of
minimize side effects and optimize safely achievable ther- pharmacogenetic therapy will be to define the optimal
apeutic levels is warranted. For example, percutaneous drug- strategy that would prevent interference with the normal

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 81 NUMBER 1 | JANUARY 2007 101


DISCOVERY

Table 1 Genetic diseases and aminoglycoside-based pharmocogenetic therapy


Affected
Disease gene Model Treatment outcome Reference
22
Ataxia–telangiectasia ATM In vitro cDNA-coupled Full-length functional protein
transcription
Cell culture
2,4,6,7,13,14,20,26
Cystic fibrosis CFTR Cell culture Full-length functional protein
Transgenic mice Amelioration of phenotype
Patients Amelioration of clinical
symptoms
23
Cystinosis CTNS Cell culture Full-length functional protein
2,7,15,17,24,34
Duchenne muscular dystrophy DMD Cell culture Variable
Transgenic mice
Patients
28
Factor VII deficiency F7 Cell culture Full-length functional protein
31
Familial atrial fibrillation KCNA5 Cell culture Full-length functional protein
30
Hailey–Hailey disease ATP2C1 Cell culture Full-length protein synthesis
27
Hemophilia F8 and F9 Patients Full-length functional protein
25
Infantile neuronal ceroid lipofuscinosis TPP1 Cell culture full-length functional protein
33
McArdle disease PYGM Patients No significant benefit
16,21
Mucopolysaccharidosis Type 1 (Hurler IDUA In vitro cDNA-coupled Full-length functional protein
syndrome) transcription
Cell culture
29
X-linked nephrogenic diabetes insipidus AVPR2 Cell culture Full-length functional protein
Transgenic mice Amelioration of phenotype

protein synthesis process. In this regard, development of new 4. Byers, P.H. Killing the messenger: new insights into nonsense-mediated
mRNA decay. J. Clin. Invest. 109, 3–6 (2002).
drugs based on deciphering the intimate structural and 5. Roberts, R.G., Gardner, R.J. & Bobrow, M. Searching for the 1 in
functional determinants of aminoglycoside interaction with 2,400,000: a review of dystrophin gene point mutations. Hum. Mutat. 4,
ribosomes, as well as the establishment of new modalities for 1–11 (1994).
6. Lukacs, G.L. & Durie, P.R. Pharmacologic approaches to correcting
targeted drug delivery are priorities. Indeed, a better under- the basic defect in cystic fibrosis. N. Engl. J. Med. 349, 1401–1404
standing of the benefits afforded by combinatorial use of (2003).
aminoglycosides with gene-of-interest-specific promoters, 7. Holbrook, J.A., Neu-Yilik, G., Hentze, M.W. & Kulozik, A.E.
Nonsense-mediated decay approaches the clinic. Nat. Genet. 36,
enhanced local application, as well as determining the most 801–808 (2004).
favorable schedule for long-term therapy will be critical for 8. Luzzatto, L., Apirion, D. & Schlessinger, D. Mechanism of action of
the future of this promising strategy for gene code repair in streptomycin in E. coli: interruption of the ribosome cycle at the
initiation of protein synthesis. Proc. Natl. Acad. Sci. USA 60, 873–880
otherwise untreatable genetic diseases. (1968).
9. Yoshizawa, S., Fourmy, D. & Puglisi, J.D. Structural origins of gentamicin
antibiotic action. EMBO J. 17, 6437–6448 (1998).
ACKNOWLEDGMENTS
10. Ogle, J.M. et al. Recognition of cognate transfer RNA by the 30S
LVZ is the recipient of the Kogod Program on Aging Career Development ribosomal subunit. Science 292, 897–902 (2001).
Award. This work was supported by grants from the National Institutes of 11. Recht, M.I., Douthwaite, S. & Puglisi, J.D. Basis for prokaryotic specificity
Health, Marriott Heart Disease Research Program, Marriott Foundation, of action of aminoglycoside antibiotics. EMBO J. 18, 3133–3138 (1999).
Ted Nash Long Life Foundation, and Ralph Wilson Medical Research 12. Burke, J.F. & Mogg, A.E. Suppression of a nonsense mutation in
Foundation. mammalian cells in vivo by the aminoglycoside antibiotics G-418 and
paromomycin. Nucleic Acids Res. 13, 6265–6272 (1985).
13. Howard, M., Frizzell, R.A. & Bedwell, D.M. Aminoglycoside antibiotics
CONFLICT OF INTEREST restore CFTR function by overcoming premature stop mutations. Nat.
The authors declared no conflict of interest. Med. 2, 467–469 (1996).
14. Bedwell, D.M. et al. Suppression of a CFTR premature stop mutation in a
& 2007 American Society for Clinical Pharmacology and Therapeutics bronchial epithelial cell line. Nat. Med. 3, 1280–1284 (1997).
15. Barton-Davis, E.R., Cordier, L., Shoturma, D.I., Leland, S.E. & Sweeney, H.L.
1. Nirenberg, M. The genetic code: Nobel lecture, http://nobelprize.org/ Aminoglycoside antibiotics restore dystrophin function to skeletal
nobel_prizes/medicine/laureates/1968/press.html (1968). muscles of mdx mice. J. Clin. Invest. 104, 375–381 (1999).
2. Kellermayer, R. Translational readthrough induction of pathogenic 16. Keeling, K.M. & Bedwell, D.M. Clinically relevant aminoglycosides can
nonsense mutations. Eur. J. Med. Genet. (2006). suppress disease-associated premature stop mutations in the IDUA and
3. Kusik, V. Mendelian Inheritance in Man: A Catalog of Human Genes and P53 cDNAs in a mammalian translation system. J. Mol. Med. 80, 367–376
Genetic Disorders, (John Hopkins University Press: Baltimore, 1998). (2002).

102 VOLUME 81 NUMBER 1 | JANUARY 2007 | www.nature.com/cpt


DISCOVERY

17. Howard, M.T. et al. Sequence specificity of aminoglycoside-induced stop 26. Wilschanski, M. et al. Gentamicin-induced correction of CFTR function in
condon readthrough: potential implications for treatment of Duchenne patients with cystic fibrosis and CFTR stop mutations. N. Engl. J. Med.
muscular dystrophy. Ann. Neurol. 48, 164–169 (2000). 349, 1433–1441 (2003).
18. Bonetti, B., Fu, L., Moon, J. & Bedwell, D.M. The efficiency of translation 27. James, P.D. et al. Aminoglycoside suppression of nonsense mutations in
termination is determined by a synergistic interplay between upstream severe hemophilia. Blood 106, 3043–3048 (2005).
and downstream sequences in Saccharomyces cerevisiae. J. Mol. Biol. 28. Pinotti, M. et al. Intracellular readthrough of nonsense mutations by
251, 334–345 (1995). aminoglycosides in coagulation factor VII. J. Thromb. Haemost. 4,
19. Namy, O., Hatin, I. & Rousset, J.P. Impact of the six nucleotides 1308–1314 (2006).
downstream of the stop codon on translation termination. EMBO Rep. 2, 29. Sangkuhl, K. et al. Aminoglycoside-mediated rescue of a disease-causing
787–793 (2001). nonsense mutation in the V2 vasopressin receptor gene in vitro and in
20. Clancy, J.P. et al. Evidence that systemic gentamicin suppresses vivo. Hum. Mol. Genet. 13, 893–903 (2004).
premature stop mutations in patients with cystic fibrosis. Am. J. Respir. 30. Kellermayer, R., Szigeti, R., Keeling, K.M., Bedekovics, T. & Bedwell, D.M.
Crit. Care Med. 163, 1683–1692 (2001). Aminoglycosides as potential pharmacogenetic agents in the treatment
21. Hein, L.K. et al. Alpha-L-iduronidase premature stop codons and of Hailey–Hailey disease. J. Invest. Dermatol. 126, 229–231 (2006).
potential read-through in mucopolysaccharidosis type I patients. J. Mol. 31. Olson, T.M. et al. Kv1.5 channelopathy due to KCNA5 loss-of-function
Biol. 338, 453–462 (2004). mutation causes human atrial fibrillation. Hum. Mol. Genet. 15,
22. Lai, C.H. et al. Correction of ATM gene function by aminoglycoside- 2185–2191 (2006).
induced read-through of premature termination codons. Proc. Natl. 32. Porgpermdee, S. et al. Expression of SPCA1 (Hailey–Hailey disease gene
Acad. Sci. USA 101, 15676–15681 (2004). product) in acantholytic dermatoses. J. Dermatol. Sci. 40, 137–140
23. Helip-Wooley, A., Park, M.A., Lemons, R.M. & Thoene, J.G. Expression of (2005).
CTNS alleles: subcellular localization and aminoglycoside correction in 33. Schroers, A. et al. Gentamicin treatment in McArdle disease: failure to
vitro. Mol. Genet. Metab. 75, 128–133 (2002). correct myophosphorylase deficiency. Neurology 66, 285–286 (2006).
24. Politano, L. et al. Gentamicin administration in Duchenne patients with 34. Wagner, K.R. et al. Gentamicin treatment of Duchenne and Becker
premature stop codon. Preliminary results. Acta Myol. 22, 15–21 (2003). muscular dystrophy due to nonsense mutations. Ann. Neurol. 49,
25. Sleat, D.E., Sohar, I., Gin, R.M. & Lobel, P. Aminoglycoside-mediated 706–711 (2001).
suppression of nonsense mutations in late infantile neuronal ceroid 35. Xi, B., Guan, F. & Lawrence, D.S. Enhanced production of functional
lipofuscinosis. Eur. J. Paediatr. Neurol. 5, 57–62 (2001). proteins from defective genes. J. Am. Chem. Soc. 126, 5660–5661 (2004).

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 81 NUMBER 1 | JANUARY 2007 103

You might also like