Dystrophin quantification: Biological and translational research implications

Karen Anthony, Virginia Arechavala-Gomeza, Laura E Taylor, Adeline Vulin, Yuuki Kaminoh, Silvia Torelli, Lucy Feng, Narinder Janghra, Gisèle Bonne, Maud Beuvin, Rita Barresi, Matt Henderson, Steven Laval, Afrodite Lourbakos, Giles Campion, Volker Straub, Thomas Voit, Caroline A Sewry, Jennifer E Morgan, Kevin M Flanigan, Francesco Muntoni, Karen Anthony, Virginia Arechavala-Gomeza, Laura E Taylor, Adeline Vulin, Yuuki Kaminoh, Silvia Torelli, Lucy Feng, Narinder Janghra, Gisèle Bonne, Maud Beuvin, Rita Barresi, Matt Henderson, Steven Laval, Afrodite Lourbakos, Giles Campion, Volker Straub, Thomas Voit, Caroline A Sewry, Jennifer E Morgan, Kevin M Flanigan, Francesco Muntoni

Abstract

Objective: We formed a multi-institution collaboration in order to compare dystrophin quantification methods, reach a consensus on the most reliable method, and report its biological significance in the context of clinical trials.

Methods: Five laboratories with expertise in dystrophin quantification performed a data-driven comparative analysis of a single reference set of normal and dystrophinopathy muscle biopsies using quantitative immunohistochemistry and Western blotting. We developed standardized protocols and assessed inter- and intralaboratory variability over a wide range of dystrophin expression levels.

Results: Results from the different laboratories were highly concordant with minimal inter- and intralaboratory variability, particularly with quantitative immunohistochemistry. There was a good level of agreement between data generated by immunohistochemistry and Western blotting, although immunohistochemistry was more sensitive. Furthermore, mean dystrophin levels determined by alternative quantitative immunohistochemistry methods were highly comparable.

Conclusions: Considering the biological function of dystrophin at the sarcolemma, our data indicate that the combined use of quantitative immunohistochemistry and Western blotting are reliable biochemical outcome measures for Duchenne muscular dystrophy clinical trials, and that standardized protocols can be comparable between competent laboratories. The methodology validated in our study will facilitate the development of experimental therapies focused on dystrophin production and their regulatory approval.

© 2014 American Academy of Neurology.

Figures

Figure 1. Inter- and intralaboratory variability of…
Figure 1. Inter- and intralaboratory variability of dystrophin quantification using immunohistochemistry
Five laboratories each quantified the level of dystrophin expression in the same 6 biopsies using a standardized immunohistochemistry protocol; data were analyzed using the Arechavala-Gomeza method. To assess interlaboratory variability, the mean ± SD for each biopsy was calculated as well as the coefficient of variation (CV). Note how this variation is higher for those samples containing less dystrophin (E and B). To assess intraassay precision within each laboratory, the mean ± SD for each laboratory per sample was calculated as well as the average CV per laboratory. Laboratories are unidentified.
Figure 2. Assessing the agreement between different…
Figure 2. Assessing the agreement between different methods of immunohistochemical dystrophin measurement
The mean data from each method were compared in a bar chart ± SD (A) and plotted with a regression line (B). The difference between the Arechavala-Gomeza and Taylor methods was plotted against their mean in a Bland-Altman plot (C) where the mean of the differences between the methods represents the bias (i.e., the value determined by one method minus the value determined by the other method) and the upper and lower 95% confidence limits represent the upper and lower limits of agreement, respectively (the difference between the 2 methods should lie within these bounds on 95% of occasions).
Figure 3. Inter- and intralaboratory variability of…
Figure 3. Inter- and intralaboratory variability of dystrophin quantification using Western blotting
Five laboratories each quantified the level of dystrophin expression in the same 6 biopsies using a standardized Western blotting protocol. To assess interlaboratory variability, the mean ± SD for each laboratory and biopsy was plotted on a bar chart and the average coefficient of variation (CV) per laboratory calculated. To assess intralaboratory variation, the mean ± SD for each laboratory per sample was calculated as well as the average CV per laboratory. Laboratories are unidentified.
Figure 4. Assessing the agreement between immunohistochemistry…
Figure 4. Assessing the agreement between immunohistochemistry and Western blotting for dystrophin quantification
The mean immunohistochemistry and Western blotting data for each biopsy were compared in a bar chart ± SD (A) and plotted with a regression line (B). The difference between the methods was plotted against their mean in a Bland-Altman plot (C) where the mean of the differences between the methods represents the bias (i.e., the value determined by one method minus the value determined by the other method) and the upper and lower 95% confidence limits represent the upper and lower limits of agreement, respectively (the difference between the 2 methods should lie within these bounds on 95% of occasions).

References

    1. Abbs S, Tuffery-Giraud S, Bakker E, Ferlini A, Sejersen T, Mueller CR. Best practice guidelines on molecular diagnostics in Duchenne/Becker muscular dystrophies. Neuromuscul Disord 2010;20:422–427.
    1. Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol 2003;2:731–740.
    1. Bushby KM, Gardner-Medwin D, Nicholson LV, et al. The clinical, genetic and dystrophin characteristics of Becker muscular dystrophy: II: correlation of phenotype with genetic and protein abnormalities. J Neurol 1993;240:105–112.
    1. Bushby KM, Gardner-Medwin D. The clinical, genetic and dystrophin characteristics of Becker muscular dystrophy: I: natural history. J Neurol 1993;240:98–104.
    1. Chelly J, Gilgenkrantz H, Lambert M, et al. Effect of dystrophin gene deletions on mRNA levels and processing in Duchenne and Becker muscular dystrophies. Cell 1990;63:1239–1248.
    1. Goemans NM, Tulinius M, van den Akker JT, et al. Systemic administration of PRO051 in Duchenne's muscular dystrophy. N Engl J Med 2011;364:1513–1522.
    1. Kinali M, Arechavala-Gomeza V, Feng L, et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol 2009;8:918–928.
    1. Arechavala-Gomeza V, Anthony K, Morgan J, Muntoni F. Antisense oligonucleotide-mediated exon skipping for Duchenne muscular dystrophy: progress and challenges. Curr Gene Ther 2012;12:152–160.
    1. Cirak S, Arechavala-Gomeza V, Guglieri M, et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet 2011;378:595–605.
    1. Cirak S, Feng L, Anthony K, et al. Restoration of the dystrophin-associated glycoprotein complex after exon skipping therapy in Duchenne muscular dystrophy. Mol Ther 2012;20:462–467.
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol 2013;74:637–647.
    1. Hirawat S, Welch EM, Elfring GL, et al. Safety, tolerability, and pharmacokinetics of PTC124, a nonaminoglycoside nonsense mutation suppressor, following single- and multiple-dose administration to healthy male and female adult volunteers. J Clin Pharmacol 2007;47:430–444.
    1. Welch EM, Barton ER, Zhuo J, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature 2007;447:87–91.
    1. Mendell JR, Campbell K, Rodino-Klapac L, et al. Dystrophin immunity in Duchenne's muscular dystrophy. N Engl J Med 2010;363:1429–1437.
    1. Arechavala-Gomeza V, Kinali M, Feng L, et al. Revertant fibres and dystrophin traces in Duchenne muscular dystrophy: implication for clinical trials. Neuromuscul Disord 2010;20:295–301.
    1. Lourbakos A, Sipkens J, Beekman C, et al. The incidence of revertant and trace dystrophin expression in muscle biopsies of Duchenne muscular dystrophy patients with different exon deletions. Neuromuscul Disord 2011;21:643.
    1. Muntoni F; Meeting Steering Committee and TREAT-NMD Network. The development of antisense oligonucleotide therapies for Duchenne muscular dystrophy: report on a TREAT-NMD workshop hosted by the European Medicines Agency (EMA), on September 25th 2009. Neuromuscul Disord 2010;20:355–362.
    1. Taylor LE, Kaminoh YJ, Rodesch CK, Flanigan KM. Quantification of dystrophin immunofluorescence in dystrophinopathy muscle specimens. Neuropathol Appl Neurobiol 2012;38:591–601.
    1. Arechavala-Gomeza V, Kinali M, Feng L, et al. Immunohistological intensity measurements as a tool to assess sarcolemma-associated protein expression. Neuropathol Appl Neurobiol 2010;36:265–274.
    1. Beekman C, Sipkens JA, Testerink J, et al. A sensitive, reproducible and objective immunofluorescence analysis method of dystrophin in individual fibers in samples from patients with Duchenne muscular dystrophy. PLoS One 2014;9:e107494.
    1. Altman DG, Bland JM. Measurement in medicine: the analysis of method comparison studies. Statistician 1983;32:307–317.
    1. European Medicines Agency. Guideline on bioanalytical method validation. Available at: . Accessed October 16, 2014.
    1. Bland JM, Altman DG. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1986;1:307–310.
    1. Bremmer-Bout M, Aartsma-Rus A, de Meijer EJ, et al. Targeted exon skipping in transgenic hDMD mice: a model for direct preclinical screening of human-specific antisense oligonucleotides. Mol Ther 2004;10:232–240.
    1. 't Hoen PA, de Meijer EJ, Boer JM, et al. Generation and characterization of transgenic mice with the full-length human DMD gene. J Biol Chem 2008;283:5899–5907.
    1. Arechavala-Gomeza V, Feng L, Morgan JE, Muntoni F. Correspondence: measuring dystrophin—faster is not necessarily better. Nat Rev Neurol 2012;8:469.
    1. Ervasti JM, Campbell KP. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol 1993;122:809–823.
    1. Vulin A, Wein N, Strandjord DM, et al. The ZZ domain of dystrophin in DMD: making sense of missense mutations. Hum Mutat 2014;35:257–264.
    1. Wells DJ, Wells KE, Asante EA, et al. Expression of human full-length and minidystrophin in transgenic mdx mice: implications for gene therapy of Duchenne muscular dystrophy. Hum Mol Genet 1995;4:1245–1250.
    1. Ervasti JM. Dystrophin, its interactions with other proteins, and implications for muscular dystrophy. Biochim Biophys Acta 2007;1772:108–117.
    1. Henderson DM, Lin AY, Thomas DD, Ervasti JM. The carboxy-terminal third of dystrophin enhances actin binding activity. J Mol Biol 2012;416:414–424.
    1. Henderson DM, Belanto JJ, Li B, Heun-Johnson H, Ervasti JM. Internal deletion compromises the stability of dystrophin. Hum Mol Genet 2011;20:2955–2963.
    1. Phelps SF, Hauser MA, Cole NM, et al. Expression of full-length and truncated dystrophin mini-genes in transgenic mdx mice. Hum Mol Genet 1995;4:1251–1258.
    1. Brown KJ, Marathi R, Fiorillo AA, et al. Accurate quantitation of dystrophin protein in human skeletal muscle using mass spectrometry. J Bioanal Biomed 2012;(suppl 7):001.

Source: PubMed

3
Abonnieren