4.7 Article

Hereditary sensory neuropathy type 1-associated deoxysphingolipids cause neurotoxicity, acute calcium handling abnormalities and mitochondrial updates dysfunction in vitro

Journal

NEUROBIOLOGY OF DISEASE
Volume 117, Issue -, Pages 1-14

Publisher

ACADEMIC PRESS INC ELSEVIER SCIENCE
DOI: 10.1016/j.nbd.2018.05.008

Keywords

Sphingolipid; Deoxysphingolipid; Neuron; Peripheral neuropathy; ES-285; Mitochondria; Endoplasmic reticulum

Categories

Funding

  1. Medical Research Council (MRC) Centre for Neuromuscular Diseases, MRC Centre Grant [G0601943]
  2. Brain Research Trust
  3. National Institutes of Neurological Diseases and Stroke and Office of Rare Diseases [U54NS065712]
  4. UCLH Charities
  5. European Community's Seventh Framework Programme (FP7) [259867]
  6. Department of Health's National Institute for Health Research Biomedical Research Centres [BRC51/NS/MR]
  7. Medical Research Council [1354730] Funding Source: researchfish
  8. MRC [MR/M003418/1] Funding Source: UKRI

Ask authors/readers for more resources

Hereditary sensory neuropathy type 1 (HSN-1) is a peripheral neuropathy most frequently caused by mutations in the SPTLCI or SPTLC2 genes, which code for two subunits of the enzyme serine palmitoyltransferase (SPT). SPT catalyzes the first step of de novo sphingolipid synthesis. Mutations in SPT result in a change in enzyme substrate specificity, which causes the production of atypical deoxysphinganine and deoxymethylsphinganine, rather than the normal enzyme product, sphinganine. Levels of these abnormal compounds are elevated in blood of HSN-1 patients and this is thought to cause the peripheral motor and sensory nerve damage that is characteristic of the disease, by a largely unresolved mechanism. In this study, we show that exogenous application of these deoxysphingoid bases causes dose- and time-dependent neurotoxicity in primary mammalian neurons, as determined by analysis of cell survival and neurite length. Acutely, deoxysphingoid base neurotoxicity manifests in abnormal Ca2+ handling by the endoplasmic reticulum (ER) and mitochondria as well as dysregulation of cell membrane store-operated Ca2+ channels. The changes in intracellular Ca2+ handling are accompanied by an early loss of mitochondrial membrane potential in deoxysphingoid base-treated motor and sensory neurons. Thus, these results suggest that exogenous deoxysphingoid base application causes neuronal mitochondrial dysfunction and Ca2+ handling deficits, which may play a critical role in the pathogenesis of HSN-1.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available