Int J Sports Med 2022; 43(13): 1084-1096
DOI: 10.1055/a-1797-7622
Review

Current Insights into Cellular Senescence and Myotoxicity Induced by Doxorubicin: The Role of Exercise and Growth Factors

Charalampos Andreou
1   Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull
,
Antonios Matsakas
1   Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull
› Author Affiliations

Abstract

Doxorubicin is an anti-neoplasmic drug that prevents DNA replication but induces senescence and cellular toxicity. Intensive research has focused on strategies to alleviate the doxorubicin-induced skeletal myotoxicity. The aim of the present review is to critically discuss the relevant scientific evidence about the role of exercise and growth factor administration and offer novel insights about newly developed-tools to combat the adverse drug reactions of doxorubicin treatment on skeletal muscle. In the first part, we discuss current data and mechanistic details on the impact of doxorubicin on skeletal myotoxicity. We next review key aspects about the role of regular exercise and the impact of growth factors, administered either pharmacologically or via genetic interventions. Future strategies such as combination of exercise and growth factor administration remain to be established to combat the pharmacologically-induced myotoxicity.



Publication History

Received: 12 December 2021

Accepted: 14 March 2022

Accepted Manuscript online:
14 March 2022

Article published online:
06 May 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 de Lima Junior EA, Yamashita AS, Pimentel GD. et al. Doxorubicin caused severe hyperglycaemia and insulin resistance, mediated by inhibition in AMPk signalling in skeletal muscle. J Cachexia Sarcopenia Muscle 2016; 7: 615-625
  • 2 Gibson NM, Quinn CJ, Pfannenstiel KB. et al. Effects of age on multidrug resistance protein expression and doxorubicin accumulation in cardiac and skeletal muscle. Xenobiotica 2014; 44: 472-479
  • 3 Gajadeera C, Willby MJ, Green KD. et al. Antimycobacterial activity of DNA intercalator inhibitors of Mycobacterium tuberculosis primase DnaG. J Antibiot (Tokyo) 2015; 68: 153-157
  • 4 Slingerland M, Guchelaar HJ, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov Today 2012; 17: 160-166
  • 5 Yu AP, Pei XM, Sin TK. et al. Acylated and unacylated ghrelin inhibit doxorubicin-induced apoptosis in skeletal muscle. Acta Physiol (Oxf) 2014; 211: 201-213
  • 6 Hiensch AE, Bolam KA, Mijwel S. et al. Doxorubicin-induced skeletal muscle atrophy: Elucidating the underlying molecular pathways. Acta Physiol (Oxf) 2020; 229: e13400
  • 7 Rivankar S. An overview of doxorubicin formulations in cancer therapy. J Cancer Res Ther 2014; 10: 853-858
  • 8 Baxter-Holland M, Dass CR. Doxorubicin, mesenchymal stem cell toxicity and antitumour activity: implications for clinical use. J Pharm Pharmacol 2018; 70: 320-327
  • 9 Huang SC, Wu JF, Saovieng S. et al. Doxorubicin inhibits muscle inflammation after eccentric exercise. J Cachexia Sarcopenia Muscle 2017; 8: 277-284
  • 10 Shrestha B, Pokhrel AR, Darsandhari S et al. Engineering streptomyces peucetius for doxorubicin and daunorubicin biosynthesis. In: Arora D, Sharma C, Jaglan S et al. (Eds.). Pharmaceuticals from Microbes: The Bioengineering Perspective. Springer Nature; 2019: 191–209
  • 11 Chen MB, Wu XY, Gu JH. et al. Activation of AMP-activated protein kinase contributes to doxorubicin-induced cell death and apoptosis in cultured myocardial H9c2 cells. Cell Biochem Biophys 2011; 60: 311-322
  • 12 Spallarossa P, Altieri P, Barisione C. et al. p38 MAPK and JNK antagonistically control senescence and cytoplasmic p16INK4A expression in doxorubicin-treated endothelial progenitor cells. PLoS One 2010; 5: e15583
  • 13 Sung JY, Lee KY, Kim JR. et al. Interaction between mTOR pathway inhibition and autophagy induction attenuates adriamycin-induced vascular smooth muscle cell senescence through decreased expressions of p53/p21/p16. Exp Gerontol 2018; 109: 51-58
  • 14 Yoon KJ, Zhang D, Kim SJ. et al. Exercise-induced AMPK activation is involved in delay of skeletal muscle senescence. Biochem Biophys Res Commun 2019; 512: 604-610
  • 15 Hulmi JJ, Nissinen TA, Rasanen M. et al. Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J Cachexia Sarcopenia Muscle 2018; 9: 417-432
  • 16 Jiao J, Demontis F. Skeletal muscle autophagy and its role in sarcopenia and organismal aging. Curr Opin Pharmacol 2017; 34: 1-6
  • 17 Yang J, Cao RY, Li Q. et al. Muscle Atrophy in Cancer. Adv Exp Med Biol 2018; 1088: 329-346
  • 18 Nissinen TA, Degerman J, Rasanen M. et al. Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016; 6: 32695
  • 19 Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009; 89: 381-410
  • 20 Powers SK, Criswell D, Lawler J. et al. Influence of exercise and fiber type on antioxidant enzyme activity in rat skeletal muscle. Am J Physiol 1994; 266: R375-R380
  • 21 Powers SK, Lawler J, Criswell D. et al. Endurance-training-induced cellular adaptations in respiratory muscles. J Appl Physiol (1985) 1990; 68: 2114-2118
  • 22 Camera DM, Smiles WJ, Hawley JA. Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 2016; 98: 131-143
  • 23 Powers SK, Duarte JA, Le Nguyen B. et al. Endurance exercise protects skeletal muscle against both doxorubicin-induced and inactivity-induced muscle wasting. Pflugers Arch 2019; 471: 441-453
  • 24 Powers SK, Bomkamp M, Ozdemir M. et al. Mechanisms of exercise-induced preconditioning in skeletal muscles. Redox Biol 2020; 35: 101462
  • 25 Smuder AJ. Exercise stimulates beneficial adaptations to diminish doxorubicin-induced cellular toxicity. Am J Physiol Regul Integr Comp Physiol 2019; 317: R662-R672
  • 26 Esaki M, Takemura G, Kosai K. et al. Treatment with an adenoviral vector encoding hepatocyte growth factor mitigates established cardiac dysfunction in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 2008; 294: H1048-H1057
  • 27 Koleini N, Nickel BE, Wang J. et al. Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway. Oncotarget 2017; 8: 87415-87430
  • 28 Lawrence WT, Norton JA, Sporn MB. et al. The reversal of an Adriamycin induced healing impairment with chemoattractants and growth factors. Ann Surg 1986; 203: 142-147
  • 29 Rasanen M, Degerman J, Nissinen TA. et al. VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 2016; 113: 13144-13149
  • 30 Harriss DJ, MacSween A, Atkinson G. Ethical Standards in Sport and Exercise Science Research: 2020 Update. Int J Sports Med 2019; 40: 813-817
  • 31 Bredahl EC, Hydock DS. Creatine Supplementation and Doxorubicin-Induced Skeletal Muscle Dysfunction: An Ex Vivo Investigation. Nutr Cancer 2017; 69: 607-615
  • 32 Hydock DS, Lien CY, Jensen BT. et al. Characterization of the effect of in vivo doxorubicin treatment on skeletal muscle function in the rat. Anticancer Res 2011; 31: 2023-2028
  • 33 Gilliam LA, Moylan JS, Callahan LA. et al. Doxorubicin causes diaphragm weakness in murine models of cancer chemotherapy. Muscle Nerve 2011; 43: 94-102
  • 34 Hayward R, Hydock D, Gibson N. et al. Tissue retention of doxorubicin and its effects on cardiac, smooth, and skeletal muscle function. J Physiol Biochem 2013; 69: 177-187
  • 35 D’Lugos AC, Fry CS, Ormsby JC. et al. Chronic doxorubicin administration impacts satellite cell and capillary abundance in a muscle-specific manner. Physiol Rep 2019; 7: e14052
  • 36 Gilliam LA, Lark DS, Reese LR. et al. Targeted overexpression of mitochondrial catalase protects against cancer chemotherapy-induced skeletal muscle dysfunction. Am J Physiol Endocrinol Metab 2016; 311: E293-E301
  • 37 Gouspillou G, Scheede-Bergdahl C, Spendiff S. et al. Anthracycline-containing chemotherapy causes long-term impairment of mitochondrial respiration and increased reactive oxygen species release in skeletal muscle. Sci Rep 2015; 5: 8717
  • 38 Gilliam LAA, Fisher-Wellman KH, Lin CT. et al. The anticancer agent doxorubicin disrupts mitochondrial energy metabolism and redox balance in skeletal muscle. Free Radic Biol Med 2013; 65: 988-996
  • 39 Tarpey MD, Amorese AJ, Balestrieri NP. et al. Doxorubicin causes lesions in the electron transport system of skeletal muscle mitochondria that are associated with a loss of contractile function. J Biol Chem 2019; 294: 19709-19722
  • 40 Min K, Kwon OS, Smuder AJ. et al. Increased mitochondrial emission of reactive oxygen species and calpain activation are required for doxorubicin-induced cardiac and skeletal muscle myopathy. J Physiol 2015; 593: 2017-2036
  • 41 Kavazis AN, Smuder AJ, Powers SK. Effects of short-term endurance exercise training on acute doxorubicin-induced FoxO transcription in cardiac and skeletal muscle. J Appl Physiol (1985) 2014; 117: 223-230
  • 42 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. J Appl Physiol (1985) 2011; 111: 1190-1198
  • 43 Smuder AJ, Kavazis AN, Hudson MB. et al. Oxidation enhances myofibrillar protein degradation via calpain and caspase-3. Free Radic Biol Med 2010; 49: 1152-1160
  • 44 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol (1985) 2011; 110: 935-942
  • 45 Sin TK, Tam BT, Yu AP. et al. Acute Treatment of Resveratrol Alleviates Doxorubicin-Induced Myotoxicity in Aged Skeletal Muscle Through SIRT1-Dependent Mechanisms. J Gerontol A Biol Sci Med Sci 2016; 71: 730-739
  • 46 Yang MY, Lin PM, Liu YC. et al. Induction of cellular senescence by doxorubicin is associated with upregulated miR-375 and induction of autophagy in K562 cells. PLoS One 2012; 7: e37205
  • 47 de Lima EA, de Sousa LGO, de STAA. et al. Aerobic exercise, but not metformin, prevents reduction of muscular performance by AMPk activation in mice on doxorubicin chemotherapy. J Cell Physiol 2018; 233: 9652-9662
  • 48 Al-Khalaf HH, Colak D, Al-Saif M. et al. p16( INK4a) positively regulates cyclin D1 and E2F1 through negative control of AUF1. PLoS One 2011; 6: e21111
  • 49 Idorn M, Thor Straten P. Exercise and cancer: from “healthy” to “therapeutic”?. Cancer Immunol Immunother 2017; 66: 667-671
  • 50 Kim JS, Galvao DA, Newton RU. et al. Exercise-induced myokines and their effect on prostate cancer. Nat Rev Urol 2021; 18: 519-542
  • 51 Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J 2015; 21: 111-116
  • 52 Perez Regalado S, Leon J, Feriche B. Therapeutic approach for digestive system cancers and potential implications of exercise under hypoxia condition: what little is known? a narrative review. J Cancer Res Clin Oncol 2022; 148: 1107-1121
  • 53 Ruiz-Casado A, Martin-Ruiz A, Perez LM. et al. Exercise and the hallmarks of cancer. Trends Cancer 2017; 3: 423-441
  • 54 Bredahl EC, Busekrus RB, Hydock DS. The combined effect of creatine and resistance training on doxorubicin-induced muscle dysfunction. Nutr Cancer 2020; 72: 939-947
  • 55 Quinn CJ, Hydock DS. Effects of endurance exercise and doxorubicin on skeletal muscle myogenic regulatory factor expression. Muscles Ligaments Tendons J 2017; 7: 418-425
  • 56 Combs AB, Hudman SL, Bonner HW. Effect of exercise stress upon the acute toxicity of adriamycin in mice. Res Commun Chem Pathol Pharmacol 1979; 23: 395-398
  • 57 Bredahl EC, Pfannenstiel KB, Quinn CJ. et al. Effects of exercise on doxorubicin-induced skeletal muscle dysfunction. Med Sci Sports Exerc 2016; 48: 1468-1473
  • 58 Dickinson JM, D’Lugos AC, Mahmood TN. et al. Exercise protects skeletal muscle during chronic doxorubicin administration. Med Sci Sports Exerc 2017; 49: 2394-2403
  • 59 Kwon I. Protective effects of endurance exercise on skeletal muscle remodeling against doxorubicin-induced myotoxicity in mice. Phys Act Nutr 2020; 24: 11-21
  • 60 Guigni BA, Fix DK, Bivona JJ. et al. Electrical stimulation prevents doxorubicin-induced atrophy and mitochondrial loss in cultured myotubes. Am J Physiol Cell Physiol 2019; 317: C1213-C1228
  • 61 Yao CX, Shi JC, Ma CX. et al. EGF protects cells against dox-induced growth arrest through activating cyclin d1 expression. J Cell Biochem 2015; 116: 1755-1765
  • 62 Koleini N, Nickel BE, Edel AL. et al. Non-mitogenic FGF2 protects cardiomyocytes from acute doxorubicin-induced toxicity independently of the protein kinase CK2/heme oxygenase-1 pathway. Cell Tissue Res 2018; 374: 607-617
  • 63 Sontag DP, Wang J, Kardami E. et al. FGF-2 and FGF-16 protect isolated perfused mouse hearts from acute doxorubicin-induced contractile dysfunction. Cardiovasc Toxicol 2013; 13: 244-253
  • 64 Wang S, Wang Y, Zhang Z. et al. Cardioprotective effects of fibroblast growth factor 21 against doxorubicin-induced toxicity via the SIRT1/LKB1/AMPK pathway. Cell Death Dis 2017; 8: e3018
  • 65 Chen L, Xia W, Hou M. Mesenchymal stem cells attenuate doxorubicininduced cellular senescence through the VEGF/Notch/TGFbeta signaling pathway in H9c2 cardiomyocytes. Int J Mol Med 2018; 42: 674-684
  • 66 Chen T, Zhou G, Zhu Q. et al. Overexpression of vascular endothelial growth factor 165 (VEGF165) protects cardiomyocytes against doxorubicin-induced apoptosis. J Chemother 2010; 22: 402-406
  • 67 Wang J, Xiang B, Dolinsky VW. et al. Cardiac fgf-16 expression supports cardiomyocyte survival and increases resistance to doxorubicin cytotoxicity. DNA Cell Biol 2018; 37: 866-877
  • 68 Bailly K, Soulet F, Leroy D. et al. Uncoupling of cell proliferation and differentiation activities of basic fibroblast growth factor. FASEB J 2000; 14: 333-344
  • 69 Jiang ZS, Srisakuldee W, Soulet F. et al. Non-angiogenic FGF-2 protects the ischemic heart from injury, in the presence or absence of reperfusion. Cardiovasc Res 2004; 62: 154-166
  • 70 Scully D, Naseem KM, Matsakas A. Platelet biology in regenerative medicine of skeletal muscle. Acta Physiol (Oxf) 2018; 223: e13071
  • 71 Stellos K, Kopf S, Paul A. et al. Platelets in regeneration. Semin Thromb Hemost 2010; 36: 175-184
  • 72 Li H, Usas A, Poddar M. et al. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8: e64923
  • 73 Scully D, Sfyri P, Verpoorten S. et al. Platelet releasate promotes skeletal myogenesis by increasing muscle stem cell commitment to differentiation and accelerates muscle regeneration following acute injury. Acta Physiol (Oxf) 2019; 225: e13207
  • 74 Scully D, Sfyri P, Wilkinson HN. et al. Optimising platelet secretomes to deliver robust tissue-specific regeneration. J Tissue Eng Regen Med 2020; 14: 82-98
  • 75 Tsai WC, Yu TY, Chang GJ. et al. Platelet-rich plasma releasate promotes regeneration and decreases inflammation and apoptosis of injured skeletal muscle. Am J Sports Med 2018; 46: 1980-1986
  • 76 Barlow J, Sfyri PP, Mitchell R. et al. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106: 700-713