Abstract

The 427-kDa protein dystrophin is expressed in striated muscle where it physically links the interior of muscle fibers to the extracellular matrix. A range of mutations in the DMD gene encoding dystrophin lead to a severe muscular dystrophy known as Duchenne (DMD) or a typically milder form known as Becker (BMD). Patients with nonsense mutations in dystrophin are specifically targeted by stop codon read-through drugs, whereas out-of-frame deletions and insertions are targeted by exon-skipping therapies. Both treatment strategies are currently in clinical trials. Dystrophin missense mutations, however, cause a wide range of phenotypic severity in patients. The molecular and cellular consequences of such mutations are not well understood, and there are no therapies specifically targeting this genotype. Here, we have modeled two representative missense mutations, L54R and L172H, causing DMD and BMD, respectively, in full-length dystrophin. In vitro, the mutation associated with the mild phenotype (L172H) caused a minor decrease in tertiary stability, whereas the L54R mutation associated with a severe phenotype had a more dramatic effect. When stably expressed in mammalian muscle cells, the mutations caused steady-state decreases in dystrophin protein levels inversely proportional to the tertiary stability and directly caused by proteasomal degradation. Both proteasome inhibitors and heat shock activators were able to increase mutant dystrophin to WT levels, establishing the new cell lines as a platform to screen for potential therapeutics personalized to patients with destabilized dystrophin.

Highlights

  • The 427-kDa protein dystrophin is expressed in striated muscle where it physically links the interior of muscle fibers to the extracellular matrix

  • One of the key components of the dystrophin– glycoprotein complex (DGC), dystrophin, is a 427-kDa structural protein composed of an N-terminal actin binding domain (ABD1), a central rod region made of 24 spectrin-like repeats that contain binding domains for several proteins [3,4,5,6], a cysteine-rich (CR) globular domain, and an intrinsically disordered C-terminal tail (CT) [7]

  • L54R was more left-shifted (Tm = 40.03 °C), suggesting either greater thermal instability or the presence of a partially unfolded state of the protein at initiation of the experiment. These differential scanning fluorimetry (DSF) results indicate that L54R and L172H cause improper folding of dystrophin, with the mutation leading to severe disease symptoms (L54R) having a greater deleterious effect

Read more

Summary

Introduction

The 427-kDa protein dystrophin is expressed in striated muscle where it physically links the interior of muscle fibers to the extracellular matrix. Flow cytometry used to detect dystrophin showed several small molecules capable of increasing the mutant protein levels, supporting the cell lines’ potential for future highthroughput screens for dystrophin stabilizers. The DSF and DLS data indicate that the DMD and BMD missense mutations cause measurable thermal instability, but do not cause the large-order aggregation more typical of protein aggregation diseases.

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