Your browser doesn't support javascript.
loading
Inhibition of motor neuron death in vitro and in vivo by a p75 neurotrophin receptor intracellular domain fragment.
Matusica, Dusan; Alfonsi, Fabienne; Turner, Bradley J; Butler, Tim J; Shepheard, Stephanie R; Rogers, Mary-Louise; Skeldal, Sune; Underwood, Clare K; Mangelsdorf, Marie; Coulson, Elizabeth J.
Affiliation
  • Matusica D; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia Department of Anatomy & Histology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia.
  • Alfonsi F; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
  • Turner BJ; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria 3051, Australia.
  • Butler TJ; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
  • Shepheard SR; Department of Human Physiology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia.
  • Rogers ML; Department of Human Physiology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia.
  • Skeldal S; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
  • Underwood CK; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
  • Mangelsdorf M; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
  • Coulson EJ; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia e.coulson@uq.edu.au.
J Cell Sci ; 129(3): 517-30, 2016 Feb 01.
Article in En | MEDLINE | ID: mdl-26503157
ABSTRACT
The p75 neurotrophin receptor (p75(NTR); also known as NGFR) can mediate neuronal apoptosis in disease or following trauma, and facilitate survival through interactions with Trk receptors. Here we tested the ability of a p75(NTR)-derived trophic cell-permeable peptide, c29, to inhibit p75(NTR)-mediated motor neuron death. Acute c29 application to axotomized motor neuron axons decreased cell death, and systemic c29 treatment of SOD1(G93A) mice, a common model of amyotrophic lateral sclerosis, resulted in increased spinal motor neuron survival mid-disease as well as delayed disease onset. Coincident with this, c29 treatment of these mice reduced the production of p75(NTR) cleavage products. Although c29 treatment inhibited mature- and pro-nerve-growth-factor-induced death of cultured motor neurons, and these ligands induced the cleavage of p75(NTR) in motor-neuron-like NSC-34 cells, there was no direct effect of c29 on p75(NTR) cleavage. Rather, c29 promoted motor neuron survival in vitro by enhancing the activation of TrkB-dependent signaling pathways, provided that low levels of brain-derived neurotrophic factor (BDNF) were present, an effect that was replicated in vivo in SOD1(G93A) mice. We conclude that the c29 peptide facilitates BDNF-dependent survival of motor neurons in vitro and in vivo.
Subject(s)
Key words

Full text: 1 Database: MEDLINE Main subject: Cell Death / Receptor, Nerve Growth Factor / Cell-Penetrating Peptides / Motor Neurons Type of study: Prognostic_studies Limits: Animals / Female / Humans / Male Language: En Year: 2016 Type: Article

Full text: 1 Database: MEDLINE Main subject: Cell Death / Receptor, Nerve Growth Factor / Cell-Penetrating Peptides / Motor Neurons Type of study: Prognostic_studies Limits: Animals / Female / Humans / Male Language: En Year: 2016 Type: Article