Muscle-Derived Proteins as Serum Biomarkers for Monitoring Disease Progression in Three Forms of Muscular Dystrophy

Peter M Burch, Oksana Pogoryelova, Richard Goldstein, Donald Bennett, Michela Guglieri, Volker Straub, Kate Bushby, Hanns Lochmüller, Carl Morris, Peter M Burch, Oksana Pogoryelova, Richard Goldstein, Donald Bennett, Michela Guglieri, Volker Straub, Kate Bushby, Hanns Lochmüller, Carl Morris

Abstract

Background: Identifying translatable, non-invasive biomarkers of muscular dystrophy that better reflect the disease pathology than those currently available would aid the development of new therapies, the monitoring of disease progression and the response to therapy.

Objective: The goal of this study was to evaluate a panel of serum protein biomarkers with the potential to specifically detect skeletal muscle injury.

Method: Serum concentrations of skeletal troponin I (sTnI), myosin light chain 3 (Myl3), fatty acid binding protein 3 (FABP3) and muscle-type creatine kinase (CKM) proteins were measured in 74 Duchenne muscular dystrophy (DMD), 38 Becker muscular dystrophy (BMD) and 49 Limb-girdle muscular dystrophy type 2B (LGMD2B) patients and 32 healthy controls.

Results: All four proteins were significantly elevated in the serum of these three muscular dystrophy patient populations when compared to healthy controls, but, interestingly, displayed different profiles depending on the type of muscular dystrophy. Additionally, the effects of patient age, ambulatory status, cardiac function and treatment status on the serum concentrations of the proteins were investigated. Statistical analysis revealed correlations between the serum concentrations and certain clinical endpoints including forced vital capacity in DMD patients and the time to walk ten meters in LGMD2B patients. Serum concentrations of these proteins were also elevated in two preclinical models of muscular dystrophy, the mdx mouse and the golden-retriever muscular dystrophy dog.

Conclusions: These proteins, therefore, are potential muscular dystrophy biomarkers for monitoring disease progression and therapeutic response in both preclinical and clinical studies.

Keywords: Muscular dystrophy; biomarker; creatine kinase; fatty acid binding protein 3; myosin light chain 3; skeletal troponin I.

Figures

Fig.1
Fig.1
Scatter plots of the serum clinical chemistry and protein serum concentrations in muscular dystrophy patients. (A) Total serum CK, AST and ALT levels and (B) serum concentrations of sTnI, Myl3, FABP3 and CKM for the DMD, BMD, LGMD2B patient groups and healthy controls are shown. The line and error bars represent the mean and standard deviation of each group.  ***P <  0.001;  ****P <  0.0001.
Fig.2
Fig.2
Scatter plots of the protein serum concentrations in DMD patients based on clinical status. Serum concentrations of sTnI, Myl3, FABP3, CKM and total CK in (A) ambulant (N = 46) and non-ambulant (N = 29) DMD patients or (B) those classified as with (N = 25) and without (N = 50) cardiomyopathy are shown. The line and error bars represent the mean and standard deviation of each group.  ****P <  0.0001.
Fig.3
Fig.3
No significant change in patient’s serum biomarker concentrations taken approximately one year apart. Serum biomarker concentrations in serial blood samples from DMD patients (N = 26). Patients were grouped by age and individual serum biomarker concentrations from the initial (circles) and follow-up samples (squares) were plotted. P values (P) were calculated by two-way ANOVA. For age groups where no P values is given then P >  0.99 for that comparison.
Fig.4
Fig.4
Correlations of protein serum concentration and clinical measures in DMD patients. A graph of (A) FVC measurement and (B) NSAA score versus the serum concentrations of sTnI, Myl3, FABP3, CKM and total serum CK for each DMD patient. The Spearman’s rank-order correlation coefficient (r), P value (P) and number of patients in the sample set (N) is shown for each analysis.
Fig.5
Fig.5
Correlations of protein serum concentrations and clinical measures in LGMD2B patients. A graph of the (A) FVC measurement and (B) the time to walk 10 m test versus serum concentrations of sTnI, Myl3, FABP3, CKM and total serum CK for each LGMD2B patients is shown. The Spearman’s correlation coefficient (r), P value (P) and number of patients in the sample set (N) is shown for each analysis.
Fig.6
Fig.6
Scatter plots of protein serum concentrations and age in DMD, BMD and LGMD2B patients. A graph of the serum concentrations of sTnI, Myl3, FABP3, CKM and total serum CK are shown versus the age of the patient for the (A) DMD (N = 61), (B) BMD (N = 38) and(C) LGMD2B (N = 49) samples.

References

    1. Flanigan KM. The muscular dystrophies. Seminars in Neurology. 2012;32(3):255–263.
    1. Mendell JR, Shilling C, Leslie ND, Flanigan KM, al-Dahhak R, Gastier-Foster J, et al. Evidence-based path to newborn screening for Duchenne muscular dystrophy. Annals of Neurology. 2012;71(3):304–313.
    1. Davies KE. Challenges in Duchenne muscular dystrophy. Neuromuscular disorders: NMD. 1997;7(8):482–486.
    1. Laval SH, Bushby KM. Limb-girdle muscular dystrophies–from genetics to molecular pathology. Neuropathology and Applied Neurobiology. 2004;30(2):91–105.
    1. Walter MC, Reilich P, Thiele S, Schessl J, Schreiber H, Reiners K, et al. Treatment of dysferlinopathy with deflazacort: A double-blind, placebo-controlled clinical trial. Orphanet Journal of Rare Diseases. 2013;8:26.
    1. Leung DG, Wagner KR. Therapeutic advances in muscular dystrophy. Annals of Neurology. 2013;74(3):404–411.
    1. McDonald CM, Henricson EK, Abresch RT, Han JJ, Escolar DM, Florence JM, et al. The cooperative international neuromuscular research group Duchenne natural history study–a longitudinal investigation in the era of glucocorticoid therapy: Design of protocol and the methods used. Muscle & Nerve. 2013;48(1):32–54.
    1. Ricotti V, Ridout DA, Scott E, Quinlivan R, Robb SA, Manzur AY, et al. Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. Journal of Neurology, Neurosurgery, and Psychiatry. 2013;84(6):698–705.
    1. Malik V, Rodino-Klapac LR, Mendell JR. Emerging drugs for Duchenne muscular dystrophy. Expert Opinion on Emerging Drugs. 2012;17(2):261–277.
    1. Bushby K, Connor E. Clinical outcome measures for trials in Duchenne muscular dystrophy: Report from International Working Group meetings. Clinical Investigation. 2011;1(9):1217–1235.
    1. Govoni A, Magri F, Brajkovic S, Zanetta C, Faravelli I, Corti S, et al. Ongoing therapeutic trials and outcome measures for Duchenne muscular dystrophy. Cellular and molecular life sciences: CMLS. 2013;70(23):4585–4602.
    1. Lynn S, Aartsma-Rus A, Bushby K, Furlong P, Goemans N, De Luca A, et al. Measuring clinical effectiveness of medicinal products for the treatment of Duchenne muscular dystrophy. Neuromuscular Disorders: NMD. 2014.
    1. Ozawa E, Hagiwara Y, Yoshida M. Creatine kinase, cell membrane and Duchenne muscular dystrophy. Molecular and Cellular Biochemistry. 1999;190(1-2):143–151.
    1. Ayoglu B, Chaouch A, Lochmuller H, Politano L, Bertini E, Spitali P, et al. Affinity proteomics within rare diseases: A BIO-NMD study for blood biomarkers of muscular dystrophies. EMBO Molecular Medicine. 2014;6(7):918–936.
    1. Gunst JJ, Langlois MR, Delanghe JR, De Buyzere ML, Leroux-Roels GG. Serum creatine kinase activity is not a reliable marker for muscle damage in conditions associated with low extracellular glutathione concentration. ClinicalChemistry. 1998;44(5):939–943.
    1. Cacchiarelli D, Legnini I, Martone J, Cazzella V, D’Amico A, Bertini E, et al. miRNAs as serum biomarkers for Duchenne muscular dystrophy. EMBO Molecular Medicine. 2011;3(5):258–265.
    1. Hu J, Kong M, Ye Y, Hong S, Cheng L, Jiang L. Serum miR-206 and other muscle-specific microRNAs as non-invasive biomarkers for Duchenne muscular dystrophy. Journal of Neurochemistry. 2014;129(5):877–883.
    1. Zaharieva IT, Calissano M, Scoto M, Preston M, Cirak S, Feng L, et al. Dystromirs as serum biomarkers for monitoring the disease severity in Duchenne muscular Dystrophy. PloS One. 2013;8(11):e80263.
    1. Li X, Li Y, Zhao L, Zhang D, Yao X, Zhang H, et al. Circulating Muscle-specific miRNAs in Duchenne Muscular Dystrophy Patients. Molecular Therapy Nucleic Acidse. 2014;3:177.
    1. Cynthia Martin F, Hiller M, Spitali P, Oonk S, Dalebout H, Palmblad M, et al. Fibronectin is a serum biomarker for Duchenne muscular dystrophy. Proteomics Clinical Applications. 2014;8(3-4):269–278.
    1. Nadarajah VD, van Putten M, Chaouch A, Garrood P, Straub V, Lochmuller H, et al. Serum matrix metalloproteinase-9 (MMP-9) as a biomarker for monitoring disease progression in Duchenne muscular dystrophy (DMD) Neuromuscular Disorders: NMD. 2011;21(8):569–578.
    1. Rouillon J, Zocevic A, Leger T, Garcia C, Camadro JM, Udd B, et al. Proteomics profiling of urine reveals specific titin fragments as biomarkers of Duchenne muscular dystrophy. Neuromuscular Disorders: NMD. 2014;24(7):563–573.
    1. Hathout Y, Marathi RL, Rayavarapu S, Zhang A, Brown KJ, Seol H, et al. Discovery of serum protein biomarkers in the mdx mouse model and cross-species comparison to Duchenne muscular dystrophy patients. Human Molecular Genetics. 2014.
    1. Dowling P, Holland A, Ohlendieck K. Mass Spectrometry-Based Identification of Muscle-Associated and Muscle-Derived Proteomic Biomarkers of Dystrophinopathies. Journal of Neuromuscular Diseases. 2014;1(1):15–40.
    1. Zatz M, Rapaport D, Vainzof M, Passos-Bueno MR, Bortolini ER, Pavanello Rde C, et al. Serum creatine-kinase (CK) and pyruvate-kinase (PK) activities in Duchenne (DMD) as compared with Becker (BMD) muscular dystrophy. Journal of the Neurological Sciences. 1991;102(2):190–196.
    1. McMillan HJ, Gregas M, Darras BT, Kang PB. Serum Transaminase Levels in Boys With Duchenne and Becker Muscular Dystrophy. Pediatricse. 2011;127(1):e132–e136.
    1. Veropalumbo C, Del Giudice E, Esposito G, Maddaluno S, Ruggiero L, Vajro P. Aminotransferases and muscular diseases: A disregarded lesson. Case reports and review of the literature. Journal of Paediatrics and Child Health. 2012;48(10):886–890.
    1. Willmann R, Possekel S, Dubach-Powell J, Meier T, Ruegg MA. Mammalian animal models for Duchenne muscular dystrophy. Neuromuscular Disorders: NMD. 2009;19(4):241–249.
    1. Spurney C, Shimizu R, Morgenroth LP, Kolski H, Gordish-Dressman H, Clemens PR. Cooperative International Neuromuscular Research Group Duchenne Natural History Study demonstrates insufficient diagnosis and treatment of cardiomyopathy in Duchenne muscular dystrophy. Muscle & Nerve. 2014;50(2):250–256.
    1. Angelini C, Peterle E. Old and new therapeutic developments in steroid treatment in Duchenne muscular dystrophy. Acta myologica: Myopathies and cardiomyopathies: Official journal of the Mediterranean Society of Myology / edited by the Gaetano Conte Academy for the Study of Striated Muscle Diseases. 2012;31(1):9–15.
    1. Phillips MF, Quinlivan RC, Edwards RH, Calverley PM. Changes in spirometry over time as a prognostic marker in patients with Duchenne muscular dystrophy. American Journal of Respiratory and Critical Care Medicine. 2001;164(12):2191–2194.
    1. Scott E, Eagle M, Mayhew A, Freeman J, Main M, Sheehan J, et al. Development of a functional assessment scale for ambulatory boys with Duchenne muscular dystrophy. Physiotherapy research international: The Journal for Researchers and Clinicians in Physical Therapy. 2012;17(2):101–109.
    1. Brooke MH, Fenichel GM, Griggs RC, Mendell JR, Moxley R, Miller JP, et al. Clinical investigation in Duchenne dystrophy: 2. Determination of the “power” of therapeutic trials based on thenatural history. Muscle & Nerve. 1983;6(2):91–103.
    1. Pernice W, Guggolz MA, Guggolz M, Beckmann R, Wais U. Amathematical analysis of creatine kinaseactivity in the course of Duchenne muscular dystrophy. Muscle &Nerve. 1986;9(4):333–340.
    1. Kobayashi K, Izawa T, Kuwamura M, Yamate J. Dysferlin and animal models for dysferlinopathy. Journal of Toxicologic Pathology. 2012;25(2):135–147.
    1. Brancaccio P, Maffulli N, Limongelli FM. Creatine kinase monitoring in sport medicine. British Medical Bulletin 81- 2007;82:209–230.
    1. Walker DB. Serum chemical biomarkers of cardiac injury for nonclinical safety testing. Toxicologic Pathology. 2006;34(1):94–104.
    1. Rosalki SB. Low serum creatine kinase activity. Clinical Chemistry. 1998;44(5):905.
    1. Florence JM, Fox PT, Planer GJ, Brooke MH. Activity, creatine kinase, and myoglobin in Duchenne muscular dystrophy: A clue to etiology? Neurology. 1985;35(5):758–761.
    1. Angelini C. The role of corticosteroids in muscular dystrophy: A critical appraisal. Muscle & Nerve. 2007;36(4):424–435.
    1. Shahrizaila N, Kinnear WJM, Wills AJ. Respiratory involvement in inherited primary muscle conditions. Journal of Neurology, Neurosurgery, and Psychiatry. 2006;77(10):1108–1115.
    1. Allen DG, Whitehead NP. Duchenne muscular dystrophy–what causes the increased membrane permeability in skeletal muscle? The International Journal of Biochemistry & Cell Biology. 2011;43(3):290–294.
    1. Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, et al. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature 423. 2003. pp. 168–172.
    1. Duguez S, Duddy W, Johnston H, Laine J, Le Bihan MC, Brown KJ, et al. Dystrophin deficiency leads to disturbance of LAMP1-vesicle-associated protein secretion. Cellular and Molecular Life Sciences: CMLS. 2013;70(12):2159–2174.

Source: PubMed

3
Abonneren