Your browser doesn't support javascript.
loading
α-Methyl-α-phenylsuccinimide ameliorates neurodegeneration in a C. elegans model of TDP-43 proteinopathy.
Wong, Shi Quan; Pontifex, Matthew G; Phelan, Marie M; Pidathala, Chandra; Kraemer, Brian C; Barclay, Jeff W; Berry, Neil G; O'Neill, Paul M; Burgoyne, Robert D; Morgan, Alan.
Afiliação
  • Wong SQ; Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. Electronic address: wongsq@liverpool.ac.uk.
  • Pontifex MG; Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. Electronic address: M.Pontifex@uea.ac.uk.
  • Phelan MM; Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK. Electronic address: Marie.Phelan@liverpool.ac.uk.
  • Pidathala C; Department of Chemistry, University of Liverpool, Liverpool, UK.
  • Kraemer BC; Geriatrics Research Education and Clinical Center, Seattle Veterans Affairs Puget Sound Health Care System, University of Washington Department of Medicine, Seattle, WA 98108, USA. Electronic address: kraemerb@uw.edu.
  • Barclay JW; Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. Electronic address: barclayj@liverpool.ac.uk.
  • Berry NG; Department of Chemistry, University of Liverpool, Liverpool, UK. Electronic address: ngberry@liverpool.ac.uk.
  • O'Neill PM; Department of Chemistry, University of Liverpool, Liverpool, UK. Electronic address: pmoneill@liverpool.ac.uk.
  • Burgoyne RD; Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. Electronic address: burgoyne@liverpool.ac.uk.
  • Morgan A; Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. Electronic address: amorgan@liverpool.ac.uk.
Neurobiol Dis ; 118: 40-54, 2018 10.
Article em En | MEDLINE | ID: mdl-29940336
ABSTRACT
The antiepileptic drug ethosuximide has recently been shown to be neuroprotective in various Caenorhabditis elegans and rodent neurodegeneration models. It is therefore a promising repurposing candidate for the treatment of multiple neurodegenerative diseases. However, high concentrations of the drug are required for its protective effects in animal models, which may impact on its translational potential and impede the identification of its molecular mechanism of action. Therefore, we set out to develop more potent neuroprotective lead compounds based on ethosuximide as a starting scaffold. Chemoinformatic approaches were used to identify compounds with structural similarity to ethosuximide and to prioritise these based on good predicated blood-brain barrier permeability and C. elegans bioaccumulation properties. Selected compounds were initially screened for anti-convulsant activity in a C. elegans pentylenetetrazol-induced seizure assay, as a rapid primary readout of bioactivity; and then assessed for neuroprotective properties in a C. elegans TDP-43 proteinopathy model based on pan-neuronal expression of human A315T mutant TDP-43. The most potent compound screened, α-methyl-α-phenylsuccinimide (MPS), ameliorated the locomotion defects and extended the shortened lifespan of TDP-43 mutant worms. MPS also directly protected against neurodegeneration by reducing the number of neuronal breaks and cell body losses in GFP-labelled GABAergic motor neurons. Importantly, optimal neuroprotection was exhibited by external application of 50 µM MPS, compared to 8 mM for ethosuximide. This greater potency of MPS was not due to bioaccumulation to higher internal levels within the worm, based on 1H-nuclear magnetic resonance analysis. Like ethosuximide, the activity of MPS was abolished by mutation of the evolutionarily conserved FOXO transcription factor, daf-16, suggesting that both compounds act via the same neuroprotective pathway(s). In conclusion, we have revealed a novel neuroprotective activity of MPS that is >100-fold more potent than ethosuximide. This increased potency will facilitate future biochemical studies to identify the direct molecular target(s) of both compounds, as we have shown here that they share a common downstream DAF-16-dependent mechanism of action. Furthermore, MPS is the active metabolite of another approved antiepileptic drug, methsuximide. Therefore, methsuximide may have repurposing potential for treatment of TDP-43 proteinopathies and possibly other human neurodegenerative diseases.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Succinimidas / Proteínas de Caenorhabditis elegans / Modelos Animais de Doenças / Proteinopatias TDP-43 Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Succinimidas / Proteínas de Caenorhabditis elegans / Modelos Animais de Doenças / Proteinopatias TDP-43 Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2018 Tipo de documento: Article