Abstract

BackgroundDuchenne muscular dystrophy (DMD) is an X-linked inherited disease caused by mutations in the gene encoding dystrophin that leads to a severe and ultimately life limiting muscle-wasting condition. Recombinant adeno-associated vector (rAAV)-based gene therapy is promising, but the size of the full-length dystrophin cDNA exceeds the packaging capacity of a rAAV. Alternative or complementary strategies that could treat DMD patients are thus needed. Intracellular calcium overload due to a sarcolemma permeability to calcium (SPCa) increase is an early and critical step of the DMD pathogenesis. We assessed herein whether TRPC1 and TRPC3 calcium channels may be involved in skeletal muscle SPCa alterations and could represent therapeutic targets to treat DMD.MethodsAll experiments were conducted in the DMDmdx rat, an animal model that closely reproduces the human DMD disease. We measured the cytosolic calcium concentration ([Ca2+]c) and SPCa in EDL (Extensor Digitorum Longus) muscle fibers from age-matched WT and DMDmdx rats of 1.5 to 7 months old. TRPC1 and TRPC3 expressions were measured in the EDL muscles at both the mRNA and protein levels, by RT-qPCR, western blot and immunocytofluorescence analysis.ResultsAs expected from the malignant hyperthermia like episodes observed in several DMDmdx rats, calcium homeostasis alterations were confirmed by measurements of early increases in [Ca2+]c and SPCa in muscle fibers. TRPC3 and TRPC1 protein levels were increased in DMDmdx rats. This was observed as soon as 1.5 months of age for TRPC3 but only at 7 months of age for TRPC1. A slight but reliable shift of the TRPC3 apparent molecular weight was observed in DMDmdx rat muscles. Intracellular localization of both channels was not altered. We thus focused our attention on TRPC3. Application of Pyr10, a specific inhibitor of TRPC3, abolished the differences between SPCa values measured in WT and DMDmdx. Finally, we showed that a rAAV-microdystrophin based treatment induced a high microdystrophin expression but only partial prevention of calcium homeostasis alterations, skeletal muscle force and TRPC3 protein increase.ConclusionsAll together our results show that correcting TRPC3 channel expression and/or activity appear to be a promising approach as a single or as a rAAV-based complementary therapy to treat DMD.

Highlights

  • Duchenne muscular dystrophy (DMD) is an X-linked inherited disease affecting ~ 1:5000 male births and leading to a severe, highly debilitating and life limiting muscle-wasting condition [1]

  • All these rats typically exhibited similar myopathic lesions typical of the model [27]. These lesions included isolated hyalin fibers, small clusters of degenerative fibers associated with muscle fiber regeneration foci, centro-nucleated fibers and anisocytosis with a few inflammatory cells in a slightly increased endomysial space corresponding to mild fibrosis

  • Considering the late expression alteration of TRPC1 and the earliest one for TRPC3, and that no significant change in location change is observed for the two channels, we focused our attention on TRPC3

Read more

Summary

Introduction

Duchenne muscular dystrophy (DMD) is an X-linked inherited disease affecting ~ 1:5000 male births and leading to a severe, highly debilitating and life limiting muscle-wasting condition [1]. It is urgent to find alternative or complementary therapeutic strategies for MD-based gene therapy that could treat both DMD and BMD patients. Duchenne muscular dystrophy (DMD) is an X-linked inherited disease caused by mutations in the gene encoding dystrophin that leads to a severe and life limiting muscle-wasting condition. Results: As expected from the malignant hyperthermia like episodes observed in several DMDmdx rats, calcium homeostasis alterations were confirmed by measurements of early increases in [­Ca2+]c and SPCa in muscle fibers. A slight but reliable shift of the TRPC3 apparent molecular weight was observed in DMDmdx rat muscles Intracellular localization of both channels was not altered. We showed that a rAAV-microdystrophin based treatment induced a high micro‐ dystrophin expression but only partial prevention of calcium homeostasis alterations, skeletal muscle force and TRPC3 protein increase

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call