Effect of endurance exercise on microRNAs in myositis skeletal muscle-A randomized controlled study

Jessica F Boehler, Marshall W Hogarth, Matthew D Barberio, James S Novak, Svetlana Ghimbovschi, Kristy J Brown, Li Alemo Munters, Ingela Loell, Yi-Wen Chen, Heather Gordish-Dressman, Helene Alexanderson, Ingrid E Lundberg, Kanneboyina Nagaraju, Jessica F Boehler, Marshall W Hogarth, Matthew D Barberio, James S Novak, Svetlana Ghimbovschi, Kristy J Brown, Li Alemo Munters, Ingela Loell, Yi-Wen Chen, Heather Gordish-Dressman, Helene Alexanderson, Ingrid E Lundberg, Kanneboyina Nagaraju

Abstract

Objective: To identify changes in skeletal muscle microRNA expression after endurance exercise and associate the identified microRNAs with mRNA and protein expression to disease-specific pathways in polymyositis (PM) and dermatomyositis (DM) patients.

Methods: Following a parallel clinical trial design, patients with probable PM or DM, exercising less than once a week, and on stable medication for at least one month were randomized into two groups at Karolinska University Hospital: a 12-week endurance exercise group (n = 12) or a non-exercised control group (n = 11). Using an Affymetrix microarray, microRNA expression was determined in paired muscle biopsies taken before and after the exercise intervention from 3 patients in each group. Ingenuity pathway analysis with a microRNA target filter was used to identify microRNA transcript targets. These targets were investigated at the mRNA (microarray) and protein (mass spectrometry) levels in patients.

Results: Endurance exercise altered 39 microRNAs. The microRNAs with increased expression were predicted to target transcripts involved in inflammatory processes, metabolism, and muscle atrophy. Further, these target transcripts had an associated decrease in mRNA expression in exercised patients. In particular, a decrease in the NF-κB regulator IKBKB was associated with an increase in its target microRNA (miR-196b). At the protein level, there was an increase in mitochondrial proteins (AK3, HIBADH), which were associated with a decrease in microRNAs that were predicted to regulate their expression.

Conclusion: Improvement in disease phenotype after exercise is associated with increasing microRNAs that target and downregulate immune processes at the transcript level, as well as decreasing microRNAs that target and upregulate mitochondrial content at the protein level. Therefore, microRNAs may improve disease by decreasing immune responses and increasing mitochondrial biogenesis.

Trial registration: ClinicalTrials.gov NCT01184625.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT 2010 flow diagram.
Fig 1. CONSORT 2010 flow diagram.
The flow chart depicts the numbers of participants who were randomly assigned, exercised, and were analyzed for study.
Fig 2. Overview of the data analysis…
Fig 2. Overview of the data analysis of exercised and non-exercised myositis patients.
The above work plan was used to the identify exercised-induced microRNA interactions in myositis patients. (A) Total RNA was extracted from baseline (pre) and exercised (post) muscle biopsies from the exercise group and the control group. (B) After pre values were subtracted from post values as an internal control measure, a total of 39 microRNAs were identified. Ingenutiy Pathway Analysis (IPA) MicroRNA Target Filter identified that 8 of these microRNAs had predicted transcript targets. (C) A gene expression microarray was run to identify if any of these transcript targets were altered in the patients after exercise. (D) Expression pairing between the microRNA and mRNA data sets was performed using IPA to determine biological relevance. (E) Protein was extracted from pre and post exercised muscle for SuperSILAC mass spectrometry. Expression pairing was again performed with microRNA and protein data sets since microRNAs are known to inhibit translation.
Fig 3. Validation of miR-196b expression after…
Fig 3. Validation of miR-196b expression after exercise and its associated effect on classical NF-κB signaling.
(A) RT-qPCR validated that miR-196b was increased after exercise. For normalization, pre Ct values were first subtracted from post Ct values from both miR-196b and the housekeeper, U47. Double delta Ct method was then used to calculate the average fold change (2^-((Normalized Target-Housekeeper)-(Average(Normalized Control-Housekeeper)). (B) Western blot in pre-and post exercised skeletal muscle shows an average increase of 42% in total IκBα protein after exercise.
Fig 4. Exercise induces microRNAs that target…
Fig 4. Exercise induces microRNAs that target transcripts and proteins important for muscle and immune response.
Exercise-induced microRNAs help improve disease outcomes in myositis by modulating transcripts and proteins important for aerobic metabolism, immune response, and muscle atrophy.

References

    1. Hengstman GJ, van den Hoogen FH, van Engelen BG. Treatment of the inflammatory myopathies: update and practical recommendations. Expert opinion on pharmacotherapy. 2009;10(7):1183–90. Epub 2009/05/02. doi: .
    1. Harris-Love MO, Shrader JA, Koziol D, Pahlajani N, Jain M, Smith M, et al. Distribution and severity of weakness among patients with polymyositis, dermatomyositis and juvenile dermatomyositis. Rheumatology (Oxford). 2009;48(2):134–9. Epub 2008/12/17. doi: .
    1. Tansley SL, McHugh NJ. Myositis specific and associated autoantibodies in the diagnosis and management of juvenile and adult idiopathic inflammatory myopathies. Curr Rheumatol Rep. 2014;16(12):464 Epub 2014/12/04. doi: .
    1. Malik A, Hayat G, Kalia JS, Guzman MA. Idiopathic Inflammatory Myopathies: Clinical Approach and Management. Frontiers in neurology. 2016;7:64 Epub 2016/06/01. doi: .
    1. Braun TP, Marks DL. The regulation of muscle mass by endogenous glucocorticoids. Frontiers in physiology. 2015;6:12 Epub 2015/02/19. doi: .
    1. Kruger K, Mooren FC, Pilat C. The Immunomodulatory Effects of Physical Activity. Current pharmaceutical design. 2016;22(24):3730–48. Epub 2016/03/24. .
    1. Fentem PH. ABC of sports medicine. Benefits of exercise in health and disease. BMJ. 1994;308(6939):1291–5. Epub 1994/05/14. .
    1. Alexanderson H, Dastmalchi M, Esbjornsson-Liljedahl M, Opava CH, Lundberg IE. Benefits of intensive resistance training in patients with chronic polymyositis or dermatomyositis. Arthritis and rheumatism. 2007;57(5):768–77. Epub 2007/05/29. doi: .
    1. Nader GA, Dastmalchi M, Alexanderson H, Grundtman C, Gernapudi R, Esbjornsson M, et al. A longitudinal, integrated, clinical, histological and mRNA profiling study of resistance exercise in myositis. Mol Med. 2010;16(11–12):455–64. Epub 2010/09/03. doi: .
    1. Tieu J, Lundberg IE, Limaye V. Idiopathic inflammatory myositis. Best Pract Res Clin Rheumatol. 2016;30(1):149–68. Epub 2016/07/17. doi: .
    1. Alemo Munters L, Dastmalchi M, Katz A, Esbjornsson M, Loell I, Hanna B, et al. Improved exercise performance and increased aerobic capacity after endurance training of patients with stable polymyositis and dermatomyositis. Arthritis research & therapy. 2013;15(4):R83 Epub 2013/08/15. doi: .
    1. Munters LA, Loell I, Ossipova E, Raouf J, Dastmalchi M, Lindroos E, et al. Endurance Exercise Improves Molecular Pathways of Aerobic Metabolism in Patients with Myositis. Arthritis Rheumatol. 2016. Epub 2016/02/13. doi: .
    1. Luo X, Ranade K, Talker R, Jallal B, Shen N, Yao Y. microRNA-mediated regulation of innate immune response in rheumatic diseases. Arthritis research & therapy. 2013;15(2):210 Epub 2013/04/16. doi: .
    1. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature genetics. 2006;38(2):228–33. Epub 2005/12/29. doi: .
    1. Eisenberg I, Eran A, Nishino I, Moggio M, Lamperti C, Amato AA, et al. Distinctive patterns of microRNA expression in primary muscular disorders. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(43):17016–21. Epub 2007/10/19. doi: .
    1. Georgantas RW, Streicher K, Greenberg SA, Greenlees LM, Zhu W, Brohawn PZ, et al. Inhibition of myogenic microRNAs 1, 133, and 206 by inflammatory cytokines links inflammation and muscle degeneration in adult inflammatory myopathies. Arthritis Rheumatol. 2014;66(4):1022–33. Epub 2014/04/24. doi: .
    1. Nagaraju K, Casciola-Rosen L, Lundberg I, Rawat R, Cutting S, Thapliyal R, et al. Activation of the endoplasmic reticulum stress response in autoimmune myositis: potential role in muscle fiber damage and dysfunction. Arthritis and rheumatism. 2005;52(6):1824–35. Epub 2005/06/04. doi: .
    1. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975;292(7):344–7. Epub 1975/02/13. doi: .
    1. Bohan A, Peter JB. Polymyositis and dermatomyositis (second of two parts). N Engl J Med. 1975;292(8):403–7. Epub 1975/02/20. doi: .
    1. Dorph C, Nennesmo I, Lundberg IE. Percutaneous conchotome muscle biopsy. A useful diagnostic and assessment tool. The Journal of rheumatology. 2001;28(7):1591–9. Epub 2001/07/27. .
    1. Holloway KV, O'Gorman M, Woods P, Morton JP, Evans L, Cable NT, et al. Proteomic investigation of changes in human vastus lateralis muscle in response to interval-exercise training. Proteomics. 2009;9(22):5155–74. Epub 2009/10/17. doi: .
    1. Seo J, Gordish-Dressman H, Hoffman EP. An interactive power analysis tool for microarray hypothesis testing and generation. Bioinformatics. 2006;22(7):808–14. Epub 2006/01/19. doi: .
    1. Geiger T, Wisniewski JR, Cox J, Zanivan S, Kruger M, Ishihama Y, et al. Use of stable isotope labeling by amino acids in cell culture as a spike-in standard in quantitative proteomics. Nature protocols. 2011;6(2):147–57. Epub 2011/02/05. doi: .
    1. Jorgensen SB, Richter EA, Wojtaszewski JF. Role of AMPK in skeletal muscle metabolic regulation and adaptation in relation to exercise. J Physiol. 2006;574(Pt 1):17–31. Epub 2006/05/13. doi: .
    1. Wang P, Li CG, Qi Z, Cui D, Ding S. Acute exercise stress promotes Ref1/Nrf2 signalling and increases mitochondrial antioxidant activity in skeletal muscle. Exp Physiol. 2016;101(3):410–20. Epub 2015/12/20. doi: .
    1. Heier CR, Fiorillo AA, Chaisson E, Gordish-Dressman H, Hathout Y, Damsker JM, et al. Identification of Pathway-Specific Serum Biomarkers of Response to Glucocorticoid and Infliximab Treatment in Children with Inflammatory Bowel Disease. Clinical and translational gastroenterology. 2016;7(9):e192 Epub 2016/09/16. doi: .
    1. Famulla S, Lamers D, Hartwig S, Passlack W, Horrighs A, Cramer A, et al. Pigment epithelium-derived factor (PEDF) is one of the most abundant proteins secreted by human adipocytes and induces insulin resistance and inflammatory signaling in muscle and fat cells. Int J Obes (Lond). 2011;35(6):762–72. Epub 2010/10/13. doi: .
    1. Li H, Malhotra S, Kumar A. Nuclear factor-kappa B signaling in skeletal muscle atrophy. J Mol Med (Berl). 2008;86(10):1113–26. Epub 2008/06/25. doi: .
    1. Coggan AR, Spina RJ, King DS, Rogers MA, Brown M, Nemeth PM, et al. Skeletal muscle adaptations to endurance training in 60- to 70-yr-old men and women. J Appl Physiol (1985). 1992;72(5):1780–6. Epub 1992/05/01. .
    1. Imai T, Hirayama K, Osumi E. [Muscle histopathology and responsiveness to steroid therapy in polymyositis and dermatomyositis]. Rinsho Shinkeigaku. 1995;35(3):243–6. Epub 1995/03/01. .
    1. Lundberg I, Kratz AK, Alexanderson H, Patarroyo M. Decreased expression of interleukin-1alpha, interleukin-1beta, and cell adhesion molecules in muscle tissue following corticosteroid treatment in patients with polymyositis and dermatomyositis. Arthritis Rheum. 2000;43(2):336–48. Epub 2000/02/29. doi: .
    1. Coley W, Rayavarapu S, Nagaraju K. Role of non-immune mechanisms of muscle damage in idiopathic inflammatory myopathies. Arthritis Res Ther. 2012;14(2):209 Epub 2012/05/02. doi: .

Source: PubMed

3
Abonnere