1. Academic Validation
  2. Antisense oligonucleotide silencing of FUS expression as a therapeutic approach in amyotrophic lateral sclerosis

Antisense oligonucleotide silencing of FUS expression as a therapeutic approach in amyotrophic lateral sclerosis

  • Nat Med. 2022 Jan;28(1):104-116. doi: 10.1038/s41591-021-01615-z.
Vladislav A Korobeynikov # 1 2 Alexander K Lyashchenko # 1 2 Beatriz Blanco-Redondo # 1 3 Paymaan Jafar-Nejad 4 Neil A Shneider 5 6
Affiliations

Affiliations

  • 1 Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA.
  • 2 Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
  • 3 Rudolf-Schönheimer Institute for Biochemistry, Leipzig University, Leipzig, Germany.
  • 4 Ionis Pharmaceuticals, Carlsbad, CA, USA.
  • 5 Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA. ns327@columbia.edu.
  • 6 Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University, New York, NY, USA. ns327@columbia.edu.
  • # Contributed equally.
Abstract

Fused in sarcoma (FUS) is an RNA-binding protein that is genetically and pathologically associated with rare and aggressive forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). To explore the mechanisms by which mutant FUS causes neurodegeneration in ALS-FTD, we generated a series of FUS knock-in mouse lines that express the equivalent of ALS-associated mutant FUSP525L and FUSΔEX14 protein. In FUS mutant mice, we show progressive, age-dependent motor neuron loss as a consequence of a dose-dependent gain of toxic function, associated with the insolubility of FUS and related RNA-binding proteins. In this disease-relevant mouse model of ALS-FUS, we show that ION363, a non-allele-specific FUS antisense oligonucleotide, efficiently silences Fus and reduces postnatal levels of FUS protein in the brain and spinal cord, delaying motor neuron degeneration. In a patient with ALS with a FUSP525L mutation, we provide preliminary evidence that repeated intrathecal infusions of ION363 lower wild-type and mutant FUS levels in the central nervous system, resulting in a marked reduction in the burden of FUS aggregates that are a pathological hallmark of disease. In mouse genetic and human clinical studies, we provide evidence in support of FUS silencing as a therapeutic strategy in FUS-dependent ALS and FTD.

Figures
Products