Targeted exon skipping to correct exon duplications in the dystrophin gene

Kane L Greer, Hanns Lochmüller, Kevin Flanigan, Susan Fletcher, Steve D Wilton, Kane L Greer, Hanns Lochmüller, Kevin Flanigan, Susan Fletcher, Steve D Wilton

Abstract

Duchenne muscular dystrophy is a severe muscle-wasting disease caused by mutations in the dystrophin gene that ablate functional protein expression. Although exonic deletions are the most common Duchenne muscular dystrophy lesion, duplications account for 10-15% of reported disease-causing mutations, and exon 2 is the most commonly duplicated exon. Here, we describe the in vitro evaluation of phosphorodiamidate morpholino oligomers coupled to a cell-penetrating peptide and 2'-O-methyl phosphorothioate oligonucleotides, using three distinct strategies to reframe the dystrophin transcript in patient cells carrying an exon 2 duplication. Differences in exon-skipping efficiencies in vitro were observed between oligomer analogues of the same sequence, with the phosphorodiamidate morpholino oligomer coupled to a cell-penetrating peptide proving the most effective. Differences in exon 2 excision efficiency between normal and exon 2 duplication cells, were apparent, indicating that exon context influences oligomer-induced splice switching. Skipping of a single copy of exon 2 was induced in the cells carrying an exon 2 duplication, the simplest strategy to restore the reading frame and generate a normal dystrophin transcript. In contrast, multiexon skipping of exons 2-7 to generate a Becker muscular dystrophy-like dystrophin transcript was more challenging and could only be induced efficiently with the phosphorodiamidate morpholino oligomer chemistry.

Figures

Figure 1
Figure 1
AO strategies to restore dystrophin expression in the presence of a frame-shifting exon 2 duplication. (a) Possible outcomes resulting from targeting of exon 2 only include single exon skipping (and a resultant in-frame transcript) or excising both copies of exon 2 and relying on reinitiation of translation from exon 3 or 6. (b) Induction of multiple exon skipping targeting exons 2–7 to restore the reading frame. AO, antisense oligomer.
Figure 2
Figure 2
Schematic showing predictedSR proteins binding motifs in dystrophin exon 2, with AO annealing sites indicated. Lower case letters indicates intronic sequences, while the exon is shown in upper case. The colored bars represent different motifs for predicted SR binding proteins. The heights of the bars indicate predicted binding strength. Red: SRSF1; purple: SRSF1 (IgM-BRCA1); green: SRSF5; yellow: SRSF6. AO, antisense oligomer; SR, specific serine/arginine rich; SRSF, specific serine/arginine rich splicing factor.
Figure 3
Figure 3
AO-induced excision of dystrophin exon 2 from normal human myogenic cells and patient dystrophin myogenic cells with a duplication of exon 2, using oligomers of the 2OMe and PPMO chemistries. Myogenic cells were either transfected with 2OMe AO:cationic lipoplexes at the concentrations indicated and incubated for 24 hours, or transfected with PPMOs and incubated for 96 hours. Total RNA was extracted and nested reverse-transcriptase PCR was undertaken across exons 1–4. The normal, full length, and exon 2-deleted products are 389, 451, and 327 bp, respectively. (a) Normal human myogenic cells transfected with 2OMe; (b) exon 2 duplication cells with 2OMe; (c) normal human myogenic cells with PPMO; (d) exon 2 duplication cells with PPMO; (e) sequence chromatogram showing exon 1 being spliced to exon 3; (f) schematic indicating exonic combinations and predicted amplicon size. AO, antisense oligomer; PPMO, phosphorodiamidate morpholino oligomer, coupled to a cell-penetrating peptide; UT: amplicon from untreated exon 2 duplication cells; +ve: amplicon from untreated normal cells; −ve: no template PCR negative control; 2OMe, 2′O methyl phosphorothioate oligomers.
Figure 4
Figure 4
Induced removal of dystrophin exons 2–7 from normal human myogenic and exon 2 duplication myogenic cells using 2OMe AO and PPMO cocktails. Primary human and dystrophic myogenic cell cultures were transfected with either 2OMeAO:cationic lipoplexes at the concentrations indicated and incubated for 24 hours or transfected with PPMO conjugates targeting dystrophin exons 2–7 and left for 96 hours. Total RNA was extracted and nested RT-PCR was undertaken across exons 1–10. The normal, full length, and exon 2–7 deletion transcripts are represented by products of 1,157, 1,219, and 539 bp, respectively. (a) Normal myogenic cells treated with the 2OMe cocktail. (b) Exon 2 duplication cells treated with the 2OMe cocktail. (c) Normal myogenic cells treated with the PPMO cocktail. (d) Exon 2 duplication cells treated with the PPMO cocktail. (e) Sequence chromatogram confirming identity of 539 and 210 bp amplicons. Mixed sequencing peaks are generated downstream of exon 1 due to templates representing transcripts missing exons 2–7 and 2–9. (f) Schematic indicating induced exon-skipping product sizes. AO, antisense oligomer; PPMO, phosphorodiamidate morpholino oligomer, coupled to a cell-penetrating peptide; UT: amplicon from untreated exon 2 duplication cells; +ve: amplicon from untreated normal cells; −ve: no template PCR negative control; 2OMe, 2′O methyl phosphorothioate oligomers.

References

    1. Hoffman EP, Brown RH, Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51:919–928.
    1. Hoffman EP, Fischbeck KH, Brown RH, Johnson M, Medori R, Loike JD, et al. Characterization of dystrophin in muscle-biopsy specimens from patients with Duchenne's or Becker's muscular dystrophy. N Engl J Med. 1988;318:1363–1368.
    1. Moser H. Duchenne muscular dystrophy: pathogenetic aspects and genetic prevention. Hum Genet. 1984;66:17–40.
    1. Koenig M, Monaco AP, Kunkel LM. The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell. 1988;53:219–228.
    1. Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics. 1988;2:90–95.
    1. Schwartz M, Dunø M, Palle AL, Krag T, Vissing J. Deletion of exon 16 of the dystrophin gene is not associated with disease. Hum Mutat. 2007;28:205.
    1. Aartsma-Rus A, Janson AA, van Ommen GJ, van Deutekom JC. Antisense-induced exon skipping for duplications in Duchenne muscular dystrophy. BMC Med Genet. 2007;8:43.
    1. Mann CJ, Honeyman K, McClorey G, Fletcher S, Wilton SD. Improved antisense oligonucleotide induced exon skipping in the mdx mouse model of muscular dystrophy. J Gene Med. 2002;4:644–654.
    1. Mitrpant C, Fletcher S, Iversen PL, Wilton SD. By-passing the nonsense mutation in the 4 CV mouse model of muscular dystrophy by induced exon skipping. J Gene Med. 2009;11:46–56.
    1. Aartsma-Rus A, Van Deutekom JC, Fokkema IF, Van Ommen GJ, Den Dunnen JT. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve. 2006;34:135–144.
    1. Taylor PJ, Maroulis S, Mullan GL, Pedersen RL, Baumli A, Elakis G, et al. Measurement of the clinical utility of a combined mutation detection protocol in carriers of Duchenne and Becker muscular dystrophy. J Med Genet. 2007;44:368–372.
    1. Aartsma-Rus A, De Winter CL, Janson AA, Kaman WE, Van Ommen GJ, Den Dunnen JT, et al. Functional analysis of 114 exon-internal AONs for targeted DMD exon skipping: indication for steric hindrance of SR protein binding sites. Oligonucleotides. 2005;15:284–297.
    1. Aartsma-Rus A, Janson AA, Kaman WE, Bremmer-Bout M, den Dunnen JT, Baas F, et al. Therapeutic antisense-induced exon skipping in cultured muscle cells from six different DMD patients. Hum Mol Genet. 2003;12:907–914.
    1. Forrest S, Meloni PL, Muntoni F, Kim J, Fletcher S, Wilton SD. Personalized exon skipping strategies to address clustered non-deletion dystrophin mutations. Neuromuscul Disord. 2010;20:810–816.
    1. Gurvich OL, Maiti B, Weiss RB, Aggarwal G, Howard MT, Flanigan KM. DMD exon 1 truncating point mutations: amelioration of phenotype by alternative translation initiation in exon 6. Hum Mutat. 2009;30:633–640.
    1. Hamed S, Sutherland-Smith A, Gorospe J, Kendrick-Jones J, Hoffman E. DNA sequence analysis for structure/function and mutation studies in Becker muscular dystrophy. Clin Genet. 2005;68:69–79.
    1. Wilton SD, Fall AM, Harding PL, McClorey G, Coleman C, Fletcher S. Antisense oligonucleotide-induced exon skipping across the human dystrophin gene transcript. Mol Ther. 2007;15:1288–1296.
    1. Reiss J, Rininsland F. An explanation for the constitutive exon 9 cassette splicing of the DMD gene. Hum Mol Genet. 1994;3:295–298.
    1. Chelly J, Gilgenkrantz H, Lambert M, Hamard G, Chafey P, Récan D, et al. Effect of dystrophin gene deletions on mRNA levels and processing in Duchenne and Becker muscular dystrophies. Cell. 1990;63:1239–1248.
    1. Adams AM, Harding PL, Iversen PL, Coleman C, Fletcher S, Wilton SD. Antisense oligonucleotide induced exon skipping and the dystrophin gene transcript: cocktails and chemistries. BMC Mol Biol. 2007;8:57.
    1. Witting N, Duno M, Vissing J. Becker muscular dystrophy with widespread muscle hypertrophy and a non-sense mutation of exon 2. Neuromuscul Disord. 2013;23 1:25–28.
    1. Fall AM, Johnsen R, Honeyman K, Iversen P, Fletcher S, Wilton SD. Induction of revertant fibres in the mdx mouse using antisense oligonucleotides. Genet Vaccines Ther. 2006;4:3.
    1. Banks GB, Gregorevic P, Allen JM, Finn EE, Chamberlain JS. Functional capacity of dystrophins carrying deletions in the N-terminal actin-binding domain. Hum Mol Genet. 2007;16:2105–2113.
    1. Winnard AV, Klein CJ, Coovert DD, Prior T, Papp A, Snyder P, et al. Characterization of translational frame exception patients in Duchenne/Becker muscular dystrophy. Hum Mol Genet. 1993;2:737–744.
    1. Malhotra SB, Hart KA, Klamut HJ, Thomas NS, Bodrug SE, Burghes AH, et al. Frame-shift deletions in patients with Duchenne and Becker muscular dystrophy. Science. 1988;242:755–759.
    1. Fletcher S, Adkin CF, Meloni P, Wong B, Muntoni F, Kole R, et al. Targeted exon skipping to address “leaky” mutations in the dystrophin gene. Mol Ther Nucleic Acids. 2012;1:e48.
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, Roush K, Bird L, Lowes LP, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013;74 5:637–647.
    1. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer AR. ESEfinder: A web resource to identify exonic splicing enhancers. Nucleic Acids Res. 2003;31:3568–3571.
    1. Rando TA, Blau HM. Primary mouse myoblast purification, characterization, and transplantation for cell-mediated gene therapy. J Cell Biol. 1994;125:1275–1287.
    1. Harding PL, Fall AM, Honeyman K, Fletcher S, Wilton SD. The influence of antisense oligonucleotide length on dystrophin exon skipping. Mol Ther. 2007;15:157–166.
    1. Lattanzi L, Salvatori G, Coletta M, Sonnino C, Cusella De Angelis MG, Gioglio L, et al. High efficiency myogenic conversion of human fibroblasts by adenoviral vector-mediated MyoD gene transfer. An alternative strategy for ex vivo gene therapy of primary myopathies. J Clin Invest. 1998;101:2119–2128.

Source: PubMed

3
Abonnere