M. Mort, D. Ivanov, D. N. Cooper, and N. A. Chuzhanova, A meta-analysis of nonsense mutations causing human genetic disease, Hum. Mutat, vol.29, pp.1037-1047, 2008.

P. Gupta and Y. R. Li, Upf proteins: highly conserved factors involved in nonsense mRNA mediated decay, Mol. Biol. Rep, vol.45, pp.39-55, 2018.

E. D. Karousis, S. Nasif, and O. Muhlemann, Nonsense-mediated mRNA decay: novel mechanistic insights and biological impact, Wiley Interdiscip. Rev. RNA, vol.7, pp.661-682, 2016.

N. Hug, D. Longman, and J. F. Caceres, Mechanism and regulation of the nonsense-mediated decay pathway, Nucleic Acids Res, vol.44, pp.1483-1495, 2016.

M. W. Popp and L. E. Maquat, The dharma of nonsense-mediated mRNA decay in mammalian cells, Mol. Cells, vol.37, pp.1-8, 2014.

F. Lejeune, Nonsense-mediated mRNA decay at the crossroads of many cellular pathways, BMB Rep, vol.50, pp.175-185, 2017.

H. A. Kuzmiak and L. E. Maquat, Applying nonsense-mediated mRNA decay research to the clinic: progress and challenges, Trends Mol. Med, vol.12, pp.306-316, 2006.

C. Floquet, I. Hatin, J. P. Rousset, and L. Bidou, Statistical analysis of readthrough levels for nonsense mutations in mammalian cells reveals a major determinant of response to gentamicin, PLoS Genet, vol.8, p.1002608, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00686519

L. Han and E. M. Phizicky, A rationale for tRNA modification circuits in the anticodon loop, RNA, vol.24, pp.1277-1284, 2018.

F. Tuorto and F. Lyko, Genome recoding by tRNA modifications, Open Biol, vol.6, p.160287, 2016.

H. Grosjean and E. Westhof, An integrated, structure-and energy-based view of the genetic code, Nucleic Acids Res, vol.44, pp.8020-8040, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01412326

M. P. Guy, Defects in tRNA anticodon loop 2'-O-methylation are implicated in nonsyndromic X-linked intellectual disability due to mutations in FTSJ1, Hum. Mutat, vol.36, pp.1176-1187, 2015.

L. P. Sarin and S. A. Leidel, Modify or die?-RNA modification defects in metazoans, RNA Biol, vol.11, pp.1555-1567, 2014.

K. M. Keeling, X. Xue, G. Gunn, and D. M. Bedwell, Therapeutics based on stop codon readthrough, Annu. Rev. Genomics Hum. Genet, vol.15, pp.371-394, 2014.

H. Benhabiles, J. Jia, and F. Lejeune, Nonsense Mutation Correction in Human Diseases: An Approach for Targeted Medicine, 2016.

L. Bidou, V. Allamand, J. P. Rousset, and O. Namy, Sense from nonsense: therapies for premature stop codon diseases, Trends Mol. Med, vol.18, pp.679-688, 2012.

M. Dabrowski, Z. Bukowy-bieryllo, and E. Zietkiewicz, Advances in therapeutic use of a drug-stimulated translational readthrough of premature termination codons, Mol. Med, vol.24, p.25, 2018.

A. L. Beaudet and L. Meng, Gene-targeting pharmaceuticals for single-gene disorders, Hum. Mol. Genet, vol.25, pp.18-26, 2016.

S. Gonzalez-hilarion, Rescue of nonsense mutations by amlexanox in human cells, Orphanet J. Rare Dis, vol.7, p.58, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00783891

L. Linde, Nonsense-mediated mRNA decay affects nonsense transcript levels and governs response of cystic fibrosis patients to gentamicin, J. Clin. Invest, vol.117, pp.683-692, 2007.

E. M. Welch, PTC124 targets genetic disorders caused by nonsense mutations, Nature, vol.447, pp.87-91, 2007.

R. Kayali, Read-through compound 13 restores dystrophin expression and improves muscle function in the mdx mouse model for Duchenne muscular dystrophy, Hum. Mol. Genet, vol.21, pp.4007-4020, 2012.

C. Kuschal, J. J. Digiovanna, S. G. Khan, R. A. Gatti, and K. H. Kraemer, Repair of UV photolesions in xeroderma pigmentosum group C cells induced by translational readthrough of premature termination codons, Proc. Natl Acad. Sci. USA, vol.110, pp.19483-19488, 2013.

W. J. Friesen, The nucleoside analog clitocine is a potent and efficacious readthrough agent, RNA, vol.23, pp.567-577, 2017.

X. Xue, Synthetic aminoglycosides efficiently suppress cystic fibrosis transmembrane conductance regulator nonsense mutations and are enhanced by ivacaftor, Am. J. Respir. Cell Mol. Biol, vol.50, pp.805-816, 2014.

E. Shulman, Designer aminoglycosides that selectively inhibit cytoplasmic rather than mitochondrial ribosomes show decreased ototoxicity: a strategy for the treatment of genetic diseases, J. Biol. Chem, vol.289, pp.2318-2330, 2014.

L. Du, Nonaminoglycoside compounds induce readthrough of nonsense mutations, J. Exp. Med, vol.206, pp.2285-2297, 2009.

M. Manuvakhova, K. Keeling, and D. M. Bedwell, Aminoglycoside antibiotics mediate context-dependent suppression of termination codons in a mammalian translation system, RNA, vol.6, pp.1044-1055, 2000.

W. J. Friesen, The minor gentamicin complex component, X2, is a potent premature stop codon readthrough molecule with therapeutic potential, PLoS ONE, vol.13, p.206158, 2018.

L. S. Correa-cerro, DHCR7 nonsense mutations and characterisation of mRNA nonsense mediated decay in Smith-Lemli-Opitz syndrome, J. Med. Genet, vol.42, pp.350-357, 2005.

E. Kerem, Effectiveness of PTC124 treatment of cystic fibrosis caused by nonsense mutations: a prospective phase II trial, Lancet, vol.372, pp.719-727, 2008.

E. Kerem, Ataluren for the treatment of nonsense-mutation cystic fibrosis: a randomised, double-blind, placebo-controlled phase 3 trial, Lancet Respir. Med, vol.2, pp.539-547, 2014.

H. Benhabiles, Optimized approach for the identification of highly efficient correctors of nonsense mutations in human diseases, PLoS ONE, vol.12, p.187930, 2017.

L. Bidou, Premature stop codons involved in muscular dystrophies show a broad spectrum of readthrough efficiencies in response to gentamicin treatment, Gene Ther, vol.11, pp.619-627, 2004.

N. G. Gurskaya, A. P. Pereverzev, D. B. Staroverov, N. M. Markina, and K. A. Lukyanov, Analysis of nonsense-mediated mRNA decay at the singlecell level using two fluorescent proteins, Methods Enzymol, vol.572, pp.291-314, 2016.

J. H. Burchenal, The effects of the folic acid antagonists and 2,6-diaminopurine on neoplastic disease, with special reference to acute leukemia, Cancer, vol.4, pp.549-569, 1951.

C. Friend, Effect of 2,6-diaminopurine on virus of Russian spring summner encephalitis in tissue culture, Proc. Soc. Exp. Biol. Med, vol.78, pp.150-153, 1951.

D. M. Margolis, The use of beta-D-2,6-diaminopurine dioxolane with or without mycophenolate mofetil in drug-resistant HIV infection, AIDS, vol.21, pp.2025-2032, 2007.

D. Zouharova, Antiviral activities of 2,6-diaminopurine-based acyclic nucleoside phosphonates against herpesviruses: In vitro study results with pseudorabies virus (PrV, SuHV-1), Vet. Microbiol, vol.184, pp.84-93, 2016.

Y. Kamiya, Introduction of 2,6-diaminopurines into serinol nucleic acid improves anti-miRNA performance, Chembiochem, vol.18, pp.1917-1922, 2017.

L. Pintard, Trm7p catalyses the formation of two 2'-O-methylriboses in yeast tRNA anticodon loop, EMBO J, vol.21, pp.1811-1820, 2002.

M. P. Guy, Yeast Trm7 interacts with distinct proteins for critical modifications of the tRNAPhe anticodon loop, RNA, vol.18, pp.1921-1933, 2012.

G. Ren, Y. P. Zhao, L. Yang, and C. X. Fu, Anti-proliferative effect of clitocine from the mushroom Leucopaxillus giganteus on human cervical cancer HeLa cells by inducing apoptosis, Cancer Lett, vol.262, pp.190-200, 2008.

W. S. El-deiry, WAF1, a potential mediator of p53 tumor suppression, Cell, vol.75, pp.817-825, 1993.

H. Wang, R. M. Mohammad, J. Werdell, and P. Shekhar, p53 and protein kinase C independent induction of growth arrest and apoptosis by bryostatin 1 in a highly metastatic mammary epithelial cell line: In vitro versus in vivo activity, Int. J. Mol. Med, vol.1, pp.915-923, 1998.

A. I. Geller and A. A. Rich, UGA termination suppression tRNATrp active in rabbit reticulocytes, Nature, vol.283, pp.41-46, 1980.

C. Urban, K. Zerfass, C. Fingerhut, and H. Beier, UGA suppression by tRNACmCATrp occurs in diverse virus RNAs due to a limited influence of the codon context, Nucleic Acids Res, vol.24, pp.3424-3430, 1996.

M. Dewez, The conserved Wobble uridine tRNA thiolase Ctu1-Ctu2 is required to maintain genome integrity, Proc. Natl Acad. Sci. USA, vol.105, pp.5459-5464, 2008.

V. Marchand, F. Blanloeil-oillo, M. Helm, and Y. Motorin, Illumina-based RiboMethSeq approach for mapping of 2'-O-Me residues in RNA, Nucleic Acids Res, vol.44, p.135, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01452312

I. Prokhorova, Aminoglycoside interactions and impacts on the eukaryotic ribosome, Proc. Natl Acad. Sci. USA, vol.114, pp.10899-10908, 2017.

J. Atkinson and R. Martin, Mutations to nonsense codons in human genetic disease: implications for gene therapy by nonsense suppressor tRNAs, Nucleic Acids Res, vol.22, pp.1327-1334, 1994.

G. Weckbecker and J. G. Cory, Metabolic activation of 2,6-diaminopurine and 2,6-diaminopurine-2'-deoxyriboside to antitumor agents, Adv. Enzym. Regul, vol.28, pp.125-144, 1989.

S. Blanchet, Deciphering the reading of the genetic code by near-cognate tRNA, Proc. Natl Acad. Sci. USA, vol.115, pp.3018-3023, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02145351

T. M. Schmeing, R. M. Voorhees, A. C. Kelley, and V. Ramakrishnan, How mutations in tRNA distant from the anticodon affect the fidelity of decoding, Nat. Struct. Mol. Biol, vol.18, pp.432-436, 2011.

R. M. Voorhees, The structural basis for specific decoding of AUA by isoleucine tRNA on the ribosome, Nat. Struct. Mol. Biol, vol.20, pp.641-643, 2013.

M. Haas, European Medicines Agency review of ataluren for the treatment of ambulant patients aged 5 years and older with Duchenne muscular dystrophy resulting from a nonsense mutation in the dystrophin gene, Neuromuscul. Disord, vol.25, pp.5-13, 2015.

R. A. Hettig and J. D. Adcock, Studies on the toxicity of streptomycin for man; a preliminary report, Science, vol.103, pp.355-357, 1946.

G. J. Greenwood, Neomycin ototoxicity; report of a case, AMA Arch. Otolaryngol, vol.69, pp.390-397, 1959.

M. Moosajee, Functional rescue of REP1 following treatment with PTC124 and novel derivative PTC-414 in human choroideremia fibroblasts and the nonsense-mediated zebrafish model, Hum. Mol. Genet, vol.25, pp.3416-3431, 2016.

S. M. Rowe, Suppression of CFTR premature termination codons and rescue of CFTR protein and function by the synthetic aminoglycoside NB54, J. Mol. Med, vol.89, pp.1149-1161, 2011.

D. Wang, The designer aminoglycoside NB84 significantly reduces glycosaminoglycan accumulation associated with MPS I-H in the Idua-W392X mouse, Mol. Genet. Metab, vol.105, pp.116-125, 2012.

J. A. Engelman, MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling, Science, vol.316, pp.1039-1043, 2007.

F. Lejeune and L. E. Maquat, Immunopurification and analysis of protein and RNA components of mRNP in mammalian cells, Methods Mol. Biol, vol.257, pp.115-124, 2004.