4.6 Article

Dihydropyridine receptor (DHPR, CACNA1S) congenital myopathy

Journal

ACTA NEUROPATHOLOGICA
Volume 133, Issue 4, Pages 517-533

Publisher

SPRINGER
DOI: 10.1007/s00401-016-1656-8

Keywords

DHPR; Congenital myopathy; Excitation-contraction coupling; Triad; Centronuclear myopathy; Core myopathy; Myotubular myopathy

Funding

  1. Institut National de la Sante et de la Recherche Medicale (INSERM), Centre National de la Recherche Scientifique (CNRS), University of Strasbourg
  2. France Genomique National infrastructure
  3. Investissements d'Avenir program [ANR-10-INBS-09]
  4. Fondation Maladies Rares [ANR-10-LABX-0030-INRT, ANR-10-IDEX-0002-02]
  5. Sparks the Children's medical research charity [12KCL 01-MT]
  6. Swiss National Science Foundation [31003A-146198]
  7. Diana and Steve Marienhoff Fashion Industries Guild Endowed Fellowship in Pediatric Neuromuscular Diseases
  8. National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children NHS Foundation Trust
  9. University College London
  10. Muscular Dystrophy UK
  11. Swiss National Science Foundation (SNF) [31003A_146198] Funding Source: Swiss National Science Foundation (SNF)
  12. Muscular Dystrophy UK [RA4/924, RA4/0924] Funding Source: researchfish
  13. National Institute for Health Research [NF-SI-0515-10022] Funding Source: researchfish
  14. Rosetrees Trust [M145] Funding Source: researchfish

Ask authors/readers for more resources

Muscle contraction upon nerve stimulation relies on excitation-contraction coupling (ECC) to promote the rapid and generalized release of calcium within myofibers. In skeletal muscle, ECC is performed by the direct coupling of a voltage-gated L-type Ca2+ channel (dihydropyridine receptor; DHPR) located on the T-tubule with a Ca2+ release channel (ryanodine receptor; RYR1) on the sarcoplasmic reticulum (SR) component of the triad. Here, we characterize a novel class of congenital myopathy at the morphological, molecular, and functional levels. We describe a cohort of 11 patients from 7 families presenting with perinatal hypotonia, severe axial and generalized weakness. Ophthalmoplegia is present in four patients. The analysis of muscle biopsies demonstrated a characteristic intermyofibrillar network due to SR dilatation, internal nuclei, and areas of myofibrillar disorganization in some samples. Exome sequencing revealed ten recessive or dominant mutations in CACNA1S (Ca(v)1.1), the pore-forming subunit of DHPR in skeletal muscle. Both recessive and dominant mutations correlated with a consistent phenotype, a decrease in protein level, and with a major impairment of Ca2+ release induced by depolarization in cultured myotubes. While dominant CACNA1S mutations were previously linked to malignant hyperthermia susceptibility or hypokalemic periodic paralysis, our findings strengthen the importance of DHPR for perinatal muscle function in human. These data also highlight CACNA1S and ECC as therapeutic targets for the development of treatments that may be facilitated by the previous knowledge accumulated on DHPR.

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