AMPA GluA1-flip targeted oligonucleotide therapy reduces neonatal seizures and hyperexcitability

Glutamate-activated α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-Rs) mediate the majority of excitatory neurotransmission in brain and thus are major drug targets for diseases associated with hyperexcitability or neurotoxicity. Due to the critical nature of AMPA-Rs in normal...

Full description

Saved in:
Bibliographic Details
Published inPloS one Vol. 12; no. 2; p. e0171538
Main Authors Lykens, Nicole M, Coughlin, David J, Reddi, Jyoti M, Lutz, Gordon J, Tallent, Melanie K
Format Journal Article
LanguageEnglish
Published United States Public Library of Science 08.02.2017
Public Library of Science (PLoS)
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Glutamate-activated α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-Rs) mediate the majority of excitatory neurotransmission in brain and thus are major drug targets for diseases associated with hyperexcitability or neurotoxicity. Due to the critical nature of AMPA-Rs in normal brain function, typical AMPA-R antagonists have deleterious effects on cognition and motor function, highlighting the need for more precise modulators. A dramatic increase in the flip isoform of alternatively spliced AMPA-R GluA1 subunits occurs post-seizure in humans and animal models. GluA1-flip produces higher gain AMPA channels than GluA1-flop, increasing network excitability and seizure susceptibility. Splice modulating oligonucleotides (SMOs) bind to pre-mRNA to influence alternative splicing, a strategy that can be exploited to develop more selective drugs across therapeutic areas. We developed a novel SMO, GR1, which potently and specifically decreased GluA1-flip expression throughout the brain of neonatal mice lasting at least 60 days after single intracerebroventricular injection. GR1 treatment reduced AMPA-R mediated excitatory postsynaptic currents at hippocampal CA1 synapses, without affecting long-term potentiation or long-term depression, cellular models of memory, or impairing GluA1-dependent cognition or motor function in mice. Importantly, GR1 demonstrated anti-seizure properties and reduced post-seizure hyperexcitability in neonatal mice, highlighting its drug candidate potential for treating epilepsies and other neurological diseases involving network hyperexcitability.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Conceptualization: MKT GJL.Data curation: MKT GJL.Formal analysis: NML DJC MKT GJL.Funding acquisition: MKT GJL NML.Investigation: MKT NML DJC JMR.Methodology: MKT GJL DJC NML.Project administration: MKT GJL.Resources: MKT GJL.Supervision: MKT GJL.Validation: MKT GJL DJC NML.Visualization: MKT GJL NML DJC.Writing – original draft: GJL MKT NML.Writing – review & editing: GJL MKT NML DJC.
Competing Interests: Dr. Lutz and Tallent co-founded LifeSplice Pharma, which has exclusive licensing rights to LSP-GR1. Dr. Lykens is also currently employed at LifeSplice Pharma. Drs Tallent, Lutz, and Lykens are the inventors on composition of matter and methods of use patents for GR1. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
ISSN:1932-6203
1932-6203
DOI:10.1371/journal.pone.0171538