4.6 Article

Complete reversal of Lambert-Eaton myasthenic syndrome synaptic impairment by the combined use of a K+ channel blocker and a Ca2+ channel agonist

Journal

JOURNAL OF PHYSIOLOGY-LONDON
Volume 592, Issue 16, Pages 3687-3696

Publisher

WILEY-BLACKWELL
DOI: 10.1113/jphysiol.2014.276493

Keywords

-

Funding

  1. Achievement Rewards for College Scientists (ARCS) Foundation scholarship
  2. National Science Foundation [0844604, 1249546]
  3. National Institutes of Health [GM067082]
  4. Muscular Dystrophy Association [295271]
  5. Myasthenia Associations of Australia
  6. Beeren Foundation
  7. University of Pittsburgh Central Research Development Fund
  8. Division Of Integrative Organismal Systems
  9. Direct For Biological Sciences [1249546] Funding Source: National Science Foundation

Ask authors/readers for more resources

Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune disorder in which a significant fraction of the presynaptic P/Q-type Ca2+ channels critical to the triggering of neurotransmitter release at the neuromuscular junction (NMJ) are thought to be removed. There is no cure for LEMS, and the current most commonly used symptomatic treatment option is a potassium channel blocker [3,4-diaminopyridine (3,4-DAP)] that does not completely reverse symptoms and can have dose-limiting side-effects. We previously reported the development of a novel Ca2+ channel agonist, GV-58, as a possible alternative treatment strategy for LEMS. In this study, we tested the hypothesis that the combination of GV-58 and 3,4-DAP will elicit a supra-additive increase in neurotransmitter release at LEMS model NMJs. First, we tested GV-58 in a cell survival assay to assess potential effects on cyclin-dependent kinases (Cdks) and showed that GV-58 did not affect cell survival at the relevant concentrations for Ca2+ channel effects. Then, we examined the voltage dependence of GV-58 effects on Ca2+ channels using patch clamp techniques; this showed the effects of GV-58 to be dependent upon Ca2+ channel opening. Based on this mechanism, we predicted an interaction between 3,4-DAP and GV-58. We tested this hypothesis using a mouse passive transfer model of LEMS. Using intracellular electrophysiological ex vivo recordings, we demonstrated that a combined application of 3,4-DAP plus GV-58 had a supra-additive effect that completely reversed the deficit in neurotransmitter release magnitude at LEMS model NMJs. This reversal contrasts with the less significant improvement observed with either compound alone. Our data indicate that a combination of 3,4-DAP and GV-58 represents a promising treatment option for LEMS and potentially for other disorders of the NMJ.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available