Your browser doesn't support javascript.
loading
Splice modulating antisense oligonucleotides restore some acid-alpha-glucosidase activity in cells derived from patients with late-onset Pompe disease.
Aung-Htut, May Thandar; Ham, Kristin A; Tchan, Michel; Johnsen, Russell; Schnell, Frederick J; Fletcher, Sue; Wilton, Steve D.
Afiliação
  • Aung-Htut MT; Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, 6150, Australia.
  • Ham KA; Perron Institute for Neurological and Translational Science and Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, 6009, Australia.
  • Tchan M; Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, 6150, Australia.
  • Johnsen R; Perron Institute for Neurological and Translational Science and Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, 6009, Australia.
  • Schnell FJ; Genetic Medicine, Westmead Hospital, Sydney, 2145, Australia.
  • Fletcher S; Sydney Medical School, The University of Sydney, Sydney, 2006, Australia.
  • Wilton SD; Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, 6150, Australia.
Sci Rep ; 10(1): 6702, 2020 04 21.
Article em En | MEDLINE | ID: mdl-32317649
ABSTRACT
Pompe disease is caused by mutations in the GAA gene, resulting in deficient lysosomal acid-α-glucosidase activity in patients, and a progressive decline in mobility and respiratory function. Enzyme replacement therapy is one therapeutic option, but since not all patients respond to this treatment, alternative interventions should be considered. One GAA mutation, c.-32-13T > G, impacts upon normal exon 2 splicing and is found in two-thirds of late-onset cases. We and others have explored a therapeutic strategy using splice modulating phosphorodiamidate morpholino oligomers to enhance GAA exon 2 inclusion in the mature mRNA of patients with one c.-32-13T > G allele. We designed 20 oligomers and treated fibroblasts derived from five patients to identify an oligomer sequence that maximally increased enzyme activity in all fibroblasts. The most effective splice correcting oligomer was chosen to treat forced-myogenic cells, derived from fibroblasts from nine patients carrying the c.-32-13T > G mutation. After transfection, we show increased levels of the full-length GAA transcript, acid-α-glucosidase protein, and enzyme activity in all patients' myogenic cells, regardless of the nature of the mutation in the other allele. This data encourages the initiation of clinical trials to assess the therapeutic efficacy of this oligomer for those patients carrying the c.-32-13T > G mutation.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença de Depósito de Glicogênio Tipo II / Splicing de RNA / Oligonucleotídeos Antissenso / Alfa-Glucosidases Tipo de estudo: Observational_studies Limite: Humans Idioma: En Revista: Sci Rep Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Austrália

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença de Depósito de Glicogênio Tipo II / Splicing de RNA / Oligonucleotídeos Antissenso / Alfa-Glucosidases Tipo de estudo: Observational_studies Limite: Humans Idioma: En Revista: Sci Rep Ano de publicação: 2020 Tipo de documento: Article País de afiliação: Austrália