CC BY-NC-ND 4.0 · Sports Med Int Open 2024; 08: a22831663
DOI: 10.1055/a-2283-1663
Review

Effects of Exercise-Induced Changes in Myokine Expression on the Tumor Microenvironment

1   Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
,
Dorothea Clauss
1   Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
,
Wilhelm Bloch
1   Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
› Author Affiliations

Abstract

In this narrative review, we summarize the direct and indirect effects that myokines have on the tumor microenvironment. We took studies of various cancer types and species into account. Systematic reviews and meta-analyses that matched the search terms were also considered. We searched databases for six months. As a narrative approach was chosen, no data was analyzed or reanalyzed. The goal of this narrative review is to create an overview on the topic to identify research gaps and answer the questions as to whether myokine expression may be relevant in cancer research in regard to the tumor microenvironment. Six commonly known myokines were chosen. We found strong links between the influence exercise has on interleukin-6, oncostatin M, secreted protein acidic and rich in cysteine, and irisin in the context of tumor progression and inhibition via interactions with the tumor microenvironment. It became clear that the effects of myokines on the tumor microenvironment can vary and contribute to disease progression or regression. Interactions among myokines and immune cells must also be considered and require further investigation. To date, no study has shown a clear connection, while multiple studies suggest further investigation of the topic, similar to the effects of exercise on myokine expression.



Publication History

Received: 10 October 2023
Received: 20 February 2024

Accepted: 26 April 2024

Accepted Manuscript online:
26 April 2024

Article published online:
19 June 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

Bibliographical Record
Nadira Gunasekara, Dorothea Clauss, Wilhelm Bloch. Effects of Exercise-Induced Changes in Myokine Expression on the Tumor Microenvironment. Sports Med Int Open 2024; 08: a22831663.
DOI: 10.1055/a-2283-1663
 
  • References

  • 1 Zhang X, Li Y, Liu D. Effects of exercise on the quality of life in breast cancer patients: a systematic review of randomized controlled trials. Support Care Cancer 2019; 27: 9-21 DOI: 10.1007/s00520-018-4363-2.
  • 2 Zhi X, Xie M, Zeng Y. et al. Effects of exercise intervention on quality of life in adolescent and young adult cancer patients and survivors: A meta-analysis. Integr Cancer Ther 2019; 18: 1534735419895590 DOI: 10.1177/1534735419895590.
  • 3 Liu Y-P, Hsiao M. Exercise-induced SPARC prevents tumorigenesis of colon cancer. Gut 2013; 62: 810-811 DOI: 10.1136/gutjnl-2012-303235.
  • 4 Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev 2012; 11: CD006145 DOI: 10.1002/14651858.CD006145.pub3.
  • 5 Casla S, Hojman P, Márquez-Rodas I. et al. Running away from side effects: physical exercise as a complementary intervention for breast cancer patients. Clin Transl Oncol 2015; 17: 180-196 DOI: 10.1007/s12094-014-1184-8.
  • 6 de Lima C, Alves LE, Iagher F. et al. Anaerobic exercise reduces tumor growth, cancer cachexia and increases macrophage and lymphocyte response in Walker 256 tumor-bearing rats. Eur J Appl Physiol 2008; 104: 957-964 DOI: 10.1007/s00421-008-0849-9.
  • 7 Spiliopoulou P, Gavriatopoulou M, Kastritis E. et al. Exercise-induced changes in tumor growth via tumor immunity. Sports (Basel) 2021; 9 -46 DOI: 10.3390/sports9040046.
  • 8 Wiggins JM, Opoku-Acheampong AB, Baumfalk DR. et al. Exercise and the tumor microenvironment: Potential therapeutic implications. Exerc Sport Sci Rev 2018; 46: 56-64 DOI: 10.1249/JES.0000000000000137.
  • 9 Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci 2012; 125: 5591-5596 DOI: 10.1242/jcs.116392.
  • 10 Anderson NM, Simon MC. The tumor microenvironment. Curr Biol 2020; 30: R921-R925 DOI: 10.1016/j.cub.2020.06.081.
  • 11 Arneth B. Tumor microenvironment. Medicina 2019; 56: 15 DOI: 10.3390/medicina56010015.
  • 12 Wu T, Dai Y. Tumor microenvironment and therapeutic response. Cancer Lett 2017; 387: 61-68 DOI: 10.1016/j.canlet.2016.01.043.
  • 13 Özdemir BH, Özdemir AA. How exercise affects the development and progression of hepatocellular carcinoma by changing the biomolecular status of the tumor microenvironment. Exp Clin Transplant 2022; Online Publication Date: Mar 15 2022 DOI: 10.6002/ect.2021.0456.
  • 14 Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008; 27: 5904-5912 DOI: 10.1038/onc.2008.271.
  • 15 Eschke RK, Lampit A, Schenk A. et al. Impact of physical exercise on growth and progression of cancer in rodents-A systematic review and meta-analysis. Front Oncol 2019; 9: 35 DOI: 10.3389/fonc.2019.00035.
  • 16 Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J 2015; 21: 111-116 DOI: 10.1097/PPO.0000000000000096.
  • 17 Pedersen BK. Exercise-induced myokines and their role in chronic diseases. Brain Behav Immun 2011; 25: 811-816 DOI: 10.1016/j.bbi.2011.02.010.
  • 18 Severinsen MCK, Pedersen BK. Muscle-organ crosstalk: The emerging roles of myokines. Endocr Rev 2020; 41: 594-609 DOI: 10.1210/endrev/bnaa016.
  • 19 Ruiz JR, Sui X, Lobelo F. et al. Muscular strength and adiposity as predictors of adulthood cancer mortality in men. Cancer Epidemiol Biomarkers Prev 2009; 18: 1468-1476 DOI: 10.1158/1055-9965.EPI-08-1075.
  • 20 Pedersen L, Hojman P. Muscle-to-organ cross talk mediated by myokines. Adipocyte 2012; 1: 164-167 DOI: 10.4161/adip.20344.
  • 21 Chen W, Wang L, You W. et al. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol 2021; 236: 2393-2412 DOI: 10.1002/jcp.30033.
  • 22 Weigert C, Lehmann R, Hartwig S. et al. The secretome of the working human skeletal muscle--a promising opportunity to combat the metabolic disaster?. Proteomics Clin Appl 2014; 8: 5-18 DOI: 10.1002/prca.201300094.
  • 23 Ferrari R. Writing narrative style literature reviews. Med Writ 2015; 24: 230-235 DOI: 10.1179/2047480615Z.000000000329.
  • 24 Catoire M, Kersten S. The search for exercise factors in humans. FASEB J 2015; 29: 1615-1628 DOI: 10.1096/fj.14-263699.
  • 25 Brandt C, Pedersen BK. The role of exercise-induced myokines in muscle homeostasis and the defense against chronic diseases. J Biomed Biotechnol 2010; 2010: 520258 DOI: 10.1155/2010/520258.
  • 26 Sponder M, Campean I-A, Emich M. et al. Long-term endurance training increases serum cathepsin S and decreases IL-6 and hsCRP levels. J Sports Sci 2017; 35: 2129-2134 DOI: 10.1080/02640414.2016.1258482.
  • 27 Libardi CA, de Souza GV, Cavaglieri CR. et al. Effect of resistance, endurance, and concurrent training on TNF-α, IL-6, and CRP. Med Sci Sports Exerc 2012; 44: 50-56 DOI: 10.1249/MSS.0b013e318229d2e9.
  • 28 Prestes J, Shiguemoto G, Botero JP. et al. Effects of resistance training on resistin, leptin, cytokines, and muscle force in elderly post-menopausal women. J Sports Sci 2009; 27: 1607-1615 DOI: 10.1080/02640410903352923.
  • 29 Ostrowski K, Schjerling P, Pedersen BK. Physical activity and plasma interleukin-6 in humans--effect of intensity of exercise. Eur J Appl Physiol 2000; 83: 512-515 DOI: 10.1007/s004210000312.
  • 30 Kim J-S, Wilson RL, Taaffe DR. et al. Myokine expression and tumor-suppressive effect of serum after 12 wk of exercise in prostate cancer patients on ADT. Med Sci Sports Exerc 2022; 54: 197-205 DOI: 10.1249/MSS.0000000000002783.
  • 31 Kim J-S, Taaffe DR, Galvão DA. et al. Exercise in advanced prostate cancer elevates myokine levels and suppresses in-vitro cell growth. Prostate Cancer Prostatic Dis 2022; 25: 86-92 DOI: 10.1038/s41391-022-00504-x.
  • 32 Dobashi S, Hashimoto M, Koyama K. et al. Impact of acute resistance exercise on circulating secreted protein acidic and rich in cysteine (SPARC) levels in healthy young males: A pilot study. Sci Sports 2021; 36: 492-494 DOI: 10.1016/j.scispo.2020.12.005.
  • 33 Songsorn P, Ruffino J, Vollaard NBJ. No effect of acute and chronic supramaximal exercise on circulating levels of the myokine SPARC. Eur J Sport Sci 2017; 17: 447-452 DOI: 10.1080/17461391.2016.1266392.
  • 34 Nygaard H, Slettaløkken G, Vegge G. et al. Irisin in blood increases transiently after single sessions of intense endurance exercise and heavy strength training. PLoS One 2015; 10: e0121367 DOI: 10.1371/journal.pone.0121367.
  • 35 Colpitts BH, Rioux BV, Eadie AL. et al. Irisin response to acute moderate intensity exercise and high intensity interval training in youth of different obesity statuses: A randomized crossover trial. Physiol Rep 2022; 10: e15198 DOI: 10.14814/phy2.15198.
  • 36 Hecksteden A, Wegmann M, Steffen A. et al. Irisin and exercise training in humans - results from a randomized controlled training trial. BMC Med 2013; 11: 235 DOI: 10.1186/1741-7015-11-235.
  • 37 Arazi H, Babaei P, Moghimi M. et al. Acute effects of strength and endurance exercise on serum BDNF and IGF-1 levels in older men. BMC Geriatr 2021; 21: 50 DOI: 10.1186/s12877-020-01937-6.
  • 38 Gaitán JM, Moon HY, Stremlau M. et al. Effects of aerobic exercise training on systemic biomarkers and cognition in late middle-aged adults at risk for Alzheimer's disease. Front Endocrinol 2021; 12: 660181 DOI: 10.3389/fendo.2021.660181.
  • 39 Coelho-Júnior HJ, Gonçalves IO, Sampaio RAC. et al. Effects of combined resistance and power training on cognitive function in older women: A randomized controlled trial. Int J Environ Res Public Health 2020; 17: 3435 DOI: 10.3390/ijerph17103435.
  • 40 Scheller J, Chalaris A, Schmidt-Arras D. et al. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta 2011; 1813: 878-888 DOI: 10.1016/j.bbamcr.2011.01.034.
  • 41 Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol 2014; 26: 38-47 DOI: 10.1016/j.smim.2014.01.008.
  • 42 Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther 2006; 8: S3 DOI: 10.1186/ar1917.
  • 43 Kang S, Tanaka T, Narazaki M. et al. Targeting interleukin-6 signaling in clinic. Immunity 2019; 50: 1007-1023 DOI: 10.1016/j.immuni.2019.03.026.
  • 44 Abdulrauf SI, Edvardsen K, Ho KL. et al. Vascular endothelial growth factor expression and vascular density as prognostic markers of survival in patients with low-grade astrocytoma. J Neurosurg 1998; 88: 513-520 DOI: 10.3171/jns.1998.88.3.0513.
  • 45 Ruoslahti E. Specialization of tumour vasculature. Nat Rev Cancer 2002; 2: 83-90 DOI: 10.1038/nrc724.
  • 46 Schaaf MB, Garg AD, Agostinis P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis 2018; 9: 115 DOI: 10.1038/s41419-017-0061-0.
  • 47 Wei L-H, Kuo M-L, Chen C-A. et al. Interleukin-6 promotes cervical tumor growth by VEGF-dependent angiogenesis via a STAT3 pathway. Oncogene 2003; 22: 1517-1527 DOI: 10.1038/sj.onc.1206226.
  • 48 Kumari N, Dwarakanath BS, Das A. et al. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumor Biol 2016; 37: 11553-11572 DOI: 10.1007/s13277-016-5098-7.
  • 49 Kistner TM, Pedersen BK, Lieberman DE. Interleukin 6 as an energy allocator in muscle tissue. Nat Metab 2022; 4: 170-179 DOI: 10.1038/s42255-022-00538-4.
  • 50 Orange ST, Jordan AR, Odell A. et al. Acute aerobic exercise-conditioned serum reduces colon cancer cell proliferation in vitro through interleukin-6-induced regulation of DNA damage. Int J Cancer 2022; 151: 265-274 DOI: 10.1002/ijc.33982.
  • 51 Procházka V, Papajík T, Faber E. et al. Soluble interleukin-2 receptor level predicts survival in patients with follicular lymphoma treated with cyclophosphamide, doxorubicin, vincristine and prednisone chemotherapy in the rituximab era. Leuk Lymphoma 2014; 55: 1584-1590 DOI: 10.3109/10428194.2013.850167.
  • 52 Gu F-M, Li Q-L, Gao Q. et al. IL-17 induces AKT-dependent IL-6/JAK2/STAT3 activation and tumor progression in hepatocellular carcinoma. Mol Cancer 2011; 10: 150 DOI: 10.1186/1476-4598-10-150.
  • 53 Sehgal PB. Interleukin-6 at the host-tumor interface: STAT3 in biomolecular condensates in cancer cells. Cells 2022; 11: 1164 DOI: 10.3390/cells11071164.
  • 54 Lin Y, He Z, Ye J. et al. Progress in understanding the IL-6/STAT3 pathway in colorectal cancer. Onco Targets Ther 2020; 13: 13023-13032 DOI: 10.2147/OTT.S278013.
  • 55 Wang L, Zhang F, Cui J-Y. et al. CAFs enhance paclitaxel resistance by inducing EMT through the IL‑6/JAK2/STAT3 pathway. Oncol Rep 2018; 39: 2081-2090 DOI: 10.3892/or.2018.6311.
  • 56 Huber MA, Kraut N, Beug H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol 2005; 17: 548-558 DOI: 10.1016/j.ceb.2005.08.001.
  • 57 Wang Y, Zhou BP. Epithelial-mesenchymal transition in breast cancer progression and metastasis. Chin J Cancer 2011; 30: 603-611 DOI: 10.5732/cjc.011.10226.
  • 58 Zhang P, Sun Y, Ma L. ZEB1: at the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance. Cell Cycle 2015; 14: 481-487 DOI: 10.1080/15384101.2015.1006048.
  • 59 Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal 2014; 7: re8 DOI: 10.1126/scisignal.2005189.
  • 60 Sullivan NJ, Sasser AK, Axel AE. et al. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene 2009; 28: 2940-2947 DOI: 10.1038/onc.2009.180.
  • 61 Cheteh EH, Sarne V, Ceder S. et al. Interleukin-6 derived from cancer-associated fibroblasts attenuates the p53 response to doxorubicin in prostate cancer cells. Cell Death Discov 2020; 6: 42 DOI: 10.1038/s41420-020-0272-5.
  • 62 Ene C-V, Nicolae I, Geavlete B. et al. IL-6 Signaling link between inflammatory tumor microenvironment and prostatic tumorigenesis. Anal Cell Pathol (Amst) 2022; 2022: 5980387 DOI: 10.1155/2022/5980387.
  • 63 Bousso P, Robey E. Dynamics of CD8+ T cell priming by dendritic cells in intact lymph nodes. Nat Immunol 2003; 4: 579-585 DOI: 10.1038/ni928.
  • 64 Rundqvist H, Veliça P, Barbieri L. et al. Cytotoxic T-cells mediate exercise-induced reductions in tumor growth. Elife 2020; 9: e59996 DOI: 10.7554/eLife.59996.
  • 65 Yang R, Masters AR, Fortner KA. et al. IL-6 promotes the differentiation of a subset of naive CD8+ T cells into IL-21-producing B helper CD8+ T cells. J Exp Med 2016; 213: 2281-2291 DOI: 10.1084/jem.20160417.
  • 66 Nakagawa T, Tsuruoka M, Ogura H. et al. IL-6 positively regulates Foxp3+CD8+ T cells in vivo. Int Immunol 2010; 22: 129-139 DOI: 10.1093/intimm/dxp119.
  • 67 Hsieh C-C, Hung C-H, Chiang M. et al. Hepatic stellate cells enhance liver cancer progression by inducing myeloid-derived suppressor cells through interleukin-6 signaling. Int J Mol Sci 2019; 20: 5079 DOI: 10.3390/ijms20205079.
  • 68 Gulubova MV, Chonov DC, Ivanova KV. et al. Intratumoural expression of IL-6/STAT3, IL-17 and FOXP3 immune cells in the immunosuppressive tumour microenvironment of colorectal cancer immune cells-positive for IL-6, STAT3, IL-17 and FOXP3 and colorectal cancer development. Biotechnol Biotechnol Equip 2022; 36: 327-338 DOI: 10.1080/13102818.2022.2072765.
  • 69 Pedersen L, Idorn M, Olofsson GH. et al. Voluntary running suppresses tumor growth through epinephrine- and IL-6-dependent NK cell mobilization and redistribution. Cell Metab 2016; 23: 554-562 DOI: 10.1016/j.cmet.2016.01.011.
  • 70 Queen MM, Ryan RE, Holzer RG. et al. Breast cancer cells stimulate neutrophils to produce oncostatin M: potential implications for tumor progression. Cancer Res 2005; 65: 8896-8904 DOI: 10.1158/0008-5472.CAN-05-1734.
  • 71 Richards CD, Botelho F. Oncostatin M in the regulation of connective tissue cells and macrophages in pulmonary disease. Biomedicines 2019; 7: 95 DOI: 10.3390/biomedicines7040095.
  • 72 Won Seok Hyung W, Gon Lee S, Tae Kim K. et al. Oncostatin M, a muscle-secreted myokine, recovers high-glucose-induced impairment of Akt phosphorylation by Fos induction in hippocampal neuron cells. Neuroreport 2019; 30: 765-770 DOI: 10.1097/WNR.0000000000001271.
  • 73 Komori T, Morikawa Y. Essential roles of the cytokine oncostatin M in crosstalk between muscle fibers and immune cells in skeletal muscle after aerobic exercise. J Biol Chem 2022; 298: 102686 DOI: 10.1016/j.jbc.2022.102686.
  • 74 Masjedi A, Hajizadeh F, Beigi Dargani F. et al. Oncostatin M: A mysterious cytokine in cancers. Int Immunopharmacol 2021; 90: 107158 DOI: 10.1016/j.intimp.2020.107158.
  • 75 Caligiuri A, Gitto S, Lori G. et al. Oncostatin M: From intracellular signaling to therapeutic targets in liver cancer. Cancers (Basel) 2022; 14: 4211 DOI: 10.3390/cancers14174211.
  • 76 Karimi F, Behboudi Tabrizi L. The effects of six weeks of aerobic training on the tumor volume and oncostatin-M serum level in mice with breast cancer. J Res Med Sci 2018; 17: 105-114
  • 77 Caffarel MM, Coleman N. Oncostatin M receptor is a novel therapeutic target in cervical squamous cell carcinoma. J Pathol 2014; 232: 386-390 DOI: 10.1002/path.4305.
  • 78 Junk DJ, Bryson BL, Smigiel JM. et al. Oncostatin M promotes cancer cell plasticity through cooperative STAT3-SMAD3 signaling. Oncogene 2017; 36: 4001-4013 DOI: 10.1038/onc.2017.33.
  • 79 Fuxe J, Vincent T, Garcia de Herreros A. Transcriptional crosstalk between TGF-β and stem cell pathways in tumor cell invasion: role of EMT promoting Smad complexes. Cell Cycle 2010; 9: 2363-2374 DOI: 10.4161/cc.9.12.12050.
  • 80 Simonneau M, Frouin E, Huguier V. et al. Oncostatin M is overexpressed in skin squamous-cell carcinoma and promotes tumor progression. Oncotarget 2018; 9: 36457-36473 DOI: 10.18632/oncotarget.26355.
  • 81 Araujo AM, Abaurrea A, Azcoaga P. et al. Stromal oncostatin M cytokine promotes breast cancer progression by reprogramming the tumor microenvironment. J Clin Invest 2022; 132 DOI: 10.1172/JCI148667.
  • 82 Kim MK, Kim Y, Park S. et al. Effects of steady low-intensity exercise on high-fat diet stimulated breast cancer progression via the alteration of macrophage polarization. Integr Cancer Ther 2020; 19: 1534735420949678 DOI: 10.1177/1534735420949678.
  • 83 Porter PL, Sage EH, Lane TF. et al Distribution of SPARC in normal and neoplastic human tissue. J Histochem Cytochem 995 43: 791-800 DOI: 10.1177/43.8.7622842.
  • 84 Clark CJ, Sage EH. A prototypic matricellular protein in the tumor microenvironment--where there's SPARC, there's fire. J Cell Biochem 2008; 104: 721-732 DOI: 10.1002/jcb.21688.
  • 85 Arnold SA, Brekken RA. SPARC: a matricellular regulator of tumorigenesis. J Cell Commun Signal 2009; 3: 255-273 DOI: 10.1007/s12079-009-0072-4.
  • 86 Nagaraju GPC, Sharma D. Anti-cancer role of SPARC, an inhibitor of adipogenesis. Cancer Treat Rev 2011; 37: 559-566 DOI: 10.1016/j.ctrv.2010.12.001.
  • 87 Ledda MF, Adris S, Bravo AI. et al. Suppression of SPARC expression by antisense RNA abrogates the tumorigenicity of human melanoma cells. Nat Med 1997; 3: 171-176 DOI: 10.1038/nm0297-171.
  • 88 Said N, Frierson HF, Sanchez-Carbayo M. et al. Loss of SPARC in bladder cancer enhances carcinogenesis and progression. J Clin Invest 2013; 123: 751-766 DOI: 10.1172/JCI64782.
  • 89 Vaz J, Ansari D, Sasor A. et al. SPARC: A potential prognostic and therapeutic target in pancreatic cancer. Pancreas 2015; 44: 1024-1035 DOI: 10.1097/MPA.0000000000000409.
  • 90 Liang J, Wang H, Xiao H. et al. Relationship and prognostic significance of SPARC and VEGF protein expression in colon cancer. J Exp Clin Cancer Res 2010; 29: 71 DOI: 10.1186/1756-9966-29-71.
  • 91 Bao J-M, Dang Q, Lin C-J. et al. SPARC is a key mediator of TGF-β-induced renal cancer metastasis. J Cell Physiol 2021; 236: 1926-1938 DOI: 10.1002/jcp.29975.
  • 92 Said N, Socha MJ, Olearczyk JJ. et al. Normalization of the ovarian cancer microenvironment by SPARC. Mol Cancer Res 2007; 5: 1015-1030 DOI: 10.1158/1541-7786.MCR-07-0001.
  • 93 Boström P, Wu J, Jedrychowski MP. et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012; 481: 463-468 DOI: 10.1038/nature10777.
  • 94 Cebulski K, Nowińska K, Jablońska K. et al. Expression of irisin/FNDC5 in breast cancer. Int J Mol Sci 2022; 23: 3530 DOI: 10.3390/ijms23073530.
  • 95 Maak S, Norheim F, Drevon CA. et al. Progress and challenges in the biology of FNDC5 and irisin. Endocr Rev 2021; 42: 436-456 DOI: 10.1210/endrev/bnab003.
  • 96 Maalouf G-E, El Khoury D. Exercise-induced irisin, the fat browning myokine, as a potential anticancer agent. J Obes 2019; 2019: 6561726 DOI: 10.1155/2019/6561726.
  • 97 Tsai C-L, Pan C-Y, Tseng Y-T. et al. Acute effects of high-intensity interval training and moderate-intensity continuous exercise on BDNF and irisin levels and neurocognitive performance in late middle-aged and older adults. Behav Brain Res 2021; 413: 113472 DOI: 10.1016/j.bbr.2021.113472.
  • 98 Tsuchiya Y, Ando D, Takamatsu K. et al. Resistance exercise induces a greater irisin response than endurance exercise. Metabolism 2015; 64: 1042-1050 DOI: 10.1016/j.metabol.2015.05.010.
  • 99 Qiu S, Cai X, Sun Z. et al. Chronic exercise training and circulating irisin in adults: A meta-analysis. Sports Med 2015; 45: 1577-1588 DOI: 10.1007/s40279-014-0293-4.
  • 100 Polyzos SA, Anastasilakis AD, Efstathiadou ZA. et al. Irisin in metabolic diseases. Endocrine 2018; 59: 260-274 DOI: 10.1007/s12020-017-1476-1.
  • 101 Altay DU, Keha EE, Karagüzel E. et al. The diagnostic value of FNDC5/irisin in renal cell cancer. Int Braz J Urol 2018; 44: 734-739 DOI: 10.1590/S1677-5538.IBJU.2017.0404.
  • 102 Nowinska K, Jablonska K, Pawelczyk K. et al. Expression of irisin/FNDC5 in cancer cells and stromal fibroblasts of non-small cell lung cancer. Cancers (Basel) 2019; 11: 1538 DOI: 10.3390/cancers11101538.
  • 103 Alizadeh Zarei M, Seyed Hosseini E, Haddad Kashani H. et al. Effects of the exercise-inducible myokine irisin on proliferation and malignant properties of ovarian cancer cells through the HIF-1 α signaling pathway. Sci Rep 2023; 13: 170 DOI: 10.1038/s41598-022-26700-2.
  • 104 Gannon NP, Vaughan RA, Garcia-Smith R. et al. Effects of the exercise-inducible myokine irisin on malignant and non-malignant breast epithelial cell behavior in vitro. Int J Cancer 2015; 136: E197-E202 DOI: 10.1002/ijc.29142.
  • 105 Shim MK, Yoon HY, Lee S. et al. Caspase-3/-7-specific metabolic precursor for bioorthogonal tracking of tumor apoptosis. Sci Rep 2017; 7: 16635 DOI: 10.1038/s41598-017-16653-2.
  • 106 Dolcet X, Llobet D, Pallares J. et al. NF-kB in development and progression of human cancer. Virchows Arch 2005; 446: 475-482 DOI: 10.1007/s00428-005-1264-9.
  • 107 Yang BC, Leung PS. Irisin Is a positive regulator for ferroptosis in pancreatic cancer. Mol Ther Oncolytics 2020; 18: 457-466 DOI: 10.1016/j.omto.2020.08.002.
  • 108 Löffler D, Müller U, Scheuermann K. et al. Serum irisin levels are regulated by acute strenuous exercise. J Clin Endocrinol Metab 2015; 100: 1289-1299 DOI: 10.1210/jc.2014-2932.
  • 109 Shimizu E, Hashimoto K, Okamura N. et al. Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biological Psychiatry 2003; 54: 70-75 DOI: 10.1016/S0006-3223(03)00181-1.
  • 110 Fulgenzi G, Hong Z, Tomassoni-Ardori F. et al. Novel metabolic role for BDNF in pancreatic β-cell insulin secretion. Nat Commun 2020; 11: 1950 DOI: 10.1038/s41467-020-15833-5.
  • 111 de Lima NS, De Sousa RAL, Amorim FT. et al. Moderate-intensity continuous training and high-intensity interval training improve cognition, and BDNF levels of middle-aged overweight men. Metab Brain Dis 2022; 37: 463-471 DOI: 10.1007/s11011-021-00859-5.
  • 112 Matthews VB, Aström M-B, Chan MHS. et al. Brain-derived neurotrophic factor is produced by skeletal muscle cells in response to contraction and enhances fat oxidation via activation of AMP-activated protein kinase. Diabetologia 2009; 52: 1409-1418 DOI: 10.1007/s00125-009-1364-1.
  • 113 Malekan M, Nezamabadi SS, Samami E. et al. BDNF and its signaling in cancer. J Cancer Res Clin Oncol 2023; 149: 2621-2636 DOI: 10.1007/s00432-022-04365-8.