5,557

Heart Failure-Induced Myopathy: The Predictive Role of Altered Myokines’ Profile

Alexander E. Berezin1, MD, PhD; Alexander A. Berezin2, MD

1 Senior Consultant of Therapeutic Unit, Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye, Ukraine;
2 Internal Medicine Department, Medical Academy of Postgraduate Education, Zaporozhye, Ukraine.

Conflict-of-interest statement: The author(s) declare(s) that there is no conflict of interest regarding the publication of this paper.

Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http: //creativecommons.org/licenses/by-nc/4.0/

Correspondence to: Alexander E. Berezin, Professor, MD, PhD, Senior Consultant of Therapeutic Unit, Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye, Ukraine.
Email: aeberezin@gmail.com; dr_berezin@mail.ru
Telephone: +380612894585
ORCID: 0000-0002-0660-9082

Received: October 23, 2020
Revised: November 6, 2020
Accepted: November 10 2020
Published online: November 17, 2020

ABSTRACT

The editorial is depicted the role of altered myokines profile in heart failure (HF) patients in prediction of HF-related myopathy and unfavorable clinical events. The development and progression of the HF is closely associated with the occurrence of myopathy, which leads to fatigue, low tolerability to physical exercise and increased risk of mortality. Myokines are predominantly produced by skeletal myocytes and regulate energy homeostasis, reparation, and perfusion of both skeletal muscles and myocardium. Recent clinical studies have shown that the altered profile of these myokines is strongly associated with the transformation of single-skeletal muscle fiber myosin heavy chain isoforms, impaired muscle energy metabolism and regeneration of skeletal muscles and myocardium. Although the predictive value of myokines appears to be optimistic for HF progression, the ability of altered myokines’ profile to improve discriminative potency of contemporary predictive scores in HF requires to be wide investigated in large clinical trials.

Key words: Heart failure; Biomarkers; Myokines; Myopathy; Prediction; Outcomes

© 2020 The Authors. Published by ACT Publishing Group Ltd. All rights reserved.

Berezin AE, Berezin AA. Heart Failure-Induced Myopathy: The Predictive Role of Altered Myokines’ Profile. Journal of Cardiology and Therapy 2020; 7(1): 954-958 Available from: URL: http: //www.ghrnet.org/index.php/jct/article/view/3035

INTRODUCTION

Heart failure (HF) remains a global medical and social problem due to its high economic burden to the health-care system, decreased quality of life of the patients and poor long-term prognosis[1,2]. Moreover, HF is a leading cause of CV mortality and urgent hospitalization among adults and the elderly having established CV disease[3]. Although the total number of new cases of the HF with reduced ejection fraction (HFrEF) exhibits stable rate in the last decade, the prevalence of HF with preserved ejection fraction (HFpEF) continues to remarkably growth and currently higher than that of HFrEF[4]. Despite the difference in the presence of CV risk factors and comorbidities among patients having HFrEF and HFpEF, both phenotypes of the disease have similar outcomes[5,6].

Fatigue, low tolerability to physical exercise, declined skeletal muscle mass, muscle weakness are most common clinical features of HF regardless of its phenotype and frequently associated with the development of the HF-related myopathy, sarcopenia and even cardiac cachexia[7,8]. Indeed, the physical exercise intolerance as one of most disabling symptoms of specific skeletal muscle myopathy has been defined in the HFrEF and HFpEF[9]. The HF-related skeletal myopathy characterized by decreased the number of muscle fibers, declined muscle strength, impaired metabolism of oxidative muscle fiber types due to mitochondrial dysfunction, ongoing inflammation and oxidative stress, and altered regeneration after acute muscle injury[10,11].

Myokines are predominantly produced by skeletal myocytes and regulate energy homeostasis, reparation, and perfusion of both skeletal muscles and myocardium[12]. In physiological condition myokines ensure the molecular adaptations of skeletal muscles to physical exercise and hemodynamic supply acting as regulator of exercise intolerance[13]. Because myokines are also secreted by adipocytes the altered myokine profile is responsible for metabolic or hormonal derangements in skeletal muscles and adipose tissue and probably it could be a target for the therapy of HF. Therefore, some of them, such as irisin, myostatin, interleukin-6 (IL-6), were found to be powerful predictors for cardiac remodeling and mortality in myocardial infarction and HF[13-16]. The aim of the editorial is to summarize knowledge among altered myokines profile in patients having HF and elucidate its role in prediction of HF-related myopathy and unfavorable clinical events.

Myopathy in heart failure

The progression of both HFrEF and HFpEF is associated with a transformation of skeletal muscle fiber composition and metabolism by uncertain molecular mechanisms[17]. These underlying abnormalities effect skeletal muscle energy homeostasis, structure and function through direct impairment of mitochondrial electron transport chain activity, shifted metabolic substrate utilization, increased formation of reactive oxygen species, aberrant mitochondrial dynamics, and altered ion homeostasis, microvascular inflammation and endothelial dysfunction[18]. Yet, catabolic condition due to neurohumoral and inflammatory activation aggravates the effect of the metabolic alteration[19]. In addition, diaphragm dysfunction and inspiratory muscle weakness also contribute to the development of exercise intolerance in HF patients[20]. Finally, hypoxia, deep metabolic alteration and ischemia injury of muscles lead to loss of myocytes due to necrosis and apoptosis, altered myoblast differentiation, impaired reparation and weak perfusion[21]. The result of these processes are certain perturbation of specific hormonal and myokines signal pathways, which includes an imbalance in the production of aldosterone, adipocytokines, tumor necrosis factor (TNF)-alpha, myostatin, myonectin, decorin, IL-6, IL-8, IL-15, and irisin[22]. The altered profile of these myokines is associated with the transformation of single-skeletal muscle fiber myosin heavy chain isoforms, impaired muscle energy metabolism and regeneration, which lead to the occurrence of vicious circle and progression of HF-related myopathy (Figure 1).

Finally, muscle weakness, skeletal myopathy, muscle atrophy and cachexia are the attributive factors for HF progression and they are closely associated with an increase in CV mortality, HF hospitalization, and a decrease in the quality of life and well-being[23,24].

Figure 1 Principal pathophysiological mechanisms of the development of HF-related myopathy. Abbreviations: LVEF, left ventricular ejection fraction; IL, interleukin; GDF, growth differential factor; TNF, tumor necrosis factor; RAAS, renin-angiotensin-aldosterone system; NO, nitric oxide; NPs, natriuretic peptides; VEGF, vascular endothelial growth factor.

Figure 2 The effects of the myokines in HF. Abbreviations: HFrEF, heart failure with reduced election fraction; HFpEF, heart failure with preserved ejection fraction; FFA, free fatty acid; SPARK, secreted protein, acidic and rich in cysteine; IL, interleukin; BDNF, brain-derived neurotrophic factor.

Myokines in HF-related myopathy

The skeletal muscles enable to release a wide range of the biological active molecules with variable potencies called myokines, the prolife of which was found to be altered in HF patients[25]. Although HF-related myopathy has been considered as secondary muscle injury that was associated with low capillary perfusion[26], myokines ensure adaptive metabolic auto regulation of structure and function of both skeletal and respiratory muscles, as well as myocardium[27].

There is evidence for the fact that the wide spectrum of myokines provides controversial actions on skeletal muscle cells and mediates pleiotropic effects. Most of myokines are controlled by muscle contractility function, myogenesis, muscle hypertrophy, reparation and consequently closely regulates exercise tolerance via intracellular signal pathways including Janus 1 and 2 kinases / 3 and 5 signal transducer and activator of transcription proteins / nuclear factor Kappa B, PI3 kinase / MAP kinase pathways[28]. It is interesting that some potentially pro-inflammatory myokines, such as IL-15 and IL-8, simultaneously provide angiopoetic effects and support pro-apoptotic impact on myoblasts. It has been found interrelationship between NO-mediated cellular signaling and production of the myokines in skeletal muscle cells[28]. However, hyperemia in skeletal muscle over physical exercise was strong associated with myokines release. The role of myokines in physiological condition and HF is reported in Table.

Table 1 The role of some myokines in physiological condition and in HF.
MyokineAffiliationBiological actionReferences
Physiological conditionHF
Down-regulated in HF
DecorinProteoglycan↓ accumulation of ECM, ↑ cell differentiation, ↑ proliferation, and ↓ apoptosis↓ cardiac hypertrophy, ↑ cardiac fibrosis[29,30]
IrisinMuscle tissue-secreted peptide FNDC5↑ expenditure, ↑ oxidative metabolism, ↑ myoblast differentiation, ↑ glucose uptake,↓ tolerance to physical exercise, ↑ skeletal muscle hypotrophy[31,32]
MyonectinCTRP15↑ oxidation of free fatty acid, ↑ oxidative metabolism, ↑ myoblast differentiation, ↑ glucose uptake↑ skeletal muscle hypotrophy[33,34]
BDNFNeurotrophin family ↑ myoblast proliferation, ↑ neurogenesis, ↑ angiogenesis, ↑ vascular reparation↑ tolerance to physical exercise[35,36]
IL-15pleiotropic cytokine with structural similarity with IL-2Anabolic effect, ↓ oxidative stress↑ tolerance to physical exercise, ↑ skeletal muscle mass, ↓ WAT, ↓ apoptosis of cardiac myocytes and myoblasts[37]
Up-regulated in HF
MyostatinTGF-β superfamily↑ skeletal muscle fiber-type switches, ↓ fast myosin heavy-chain expression, ↓ differentiation of myoblasts, ↑ ubiquitin-proteasomal activity in myocytes and ILGF-PKB pathway↑ skeletal muscle hypotrophy, ↑IR, ↑ autophagy, ↑ muscle weakness, ↓ exercise tolerance[38,39]
IL-8cysteine-X-cysteine family of chemokines↓ glucose disposal, ↑ IR↓ skeletal muscle energy metabolism[40,41]
OsteonectinSPARC proteinPotential mediator of collagen deposition and extracellular matrix remodelingPredictor of poor HF outcomes, ↑ cardiac contractility and reparation at early stage, ↓ cardiac myocytes survival and vascular integrity at late stage[42]
Abbreviations: TGF-β, transforming growth factor-beta; IR, insulin resistance; ILGF-PKB, insulin-like growth factor-protein kinase B; WAT, white adipose tissue; ECM, extracellular matrix; SPARC, Secreted protein acidic and rich in cysteine.

The occurrence of myopathy in HF accompanies by cross over changes in the spectrum of the myokines. Indeed, there were found elevated serum concentrations of myostatin and IL-8, whereas irisin and myonectin demonstrated significant decrease in their circulating levels. Finally, myokines influence not just skeletal muscles, but also myocardium and adipose tissue and ensure their autocrine metabolic regulation of energy homeostasis, hypertrophy, reparation, and as well as adaptation of skeletal muscles to physical exercise.

Predictive value of myokines in HF

Several myokines were able to predict adverse clinical outcomes in HF. Indeed, elevated serum irisin levels were independently associated with increased mortality and a risk of cardiac cachexia in acute and chronic HF patients respectively[43,44]. Myostatin was found an independent predictor of mortality in HF patients and re-hospitalization due to HF progression[45]. Circulating levels of BDNF were inversely associated with adverse outcomes in acute and chronic HF patients[36,46]. In addition, SPARC proteins have demonstrated an independent association with poor long-term HF-related outcomes including death, and a risk for recurrent hospitalization due to[47]. However, the exact molecular pathways, which link myokines with the HF outcomes remains uncertain and requires being clear elucidated in the future.

Conclusion: Myokines being autocrine / paracrine regulators of metabolic homeostasis of myocardium and skeletal muscles are involved in the pathogenesis of HF. Altered myokines’ profile has been found in patients with HFrEF and HFpEF and was associated with poor clinical outcomes, adverse cardiac remodeling, HF-related myopathy and cardiac cachexia. Predictive value of myokines continues to investigate because preliminary findings appear to be optimistic for creation of predictive models. Large clinical trials are required to thoroughly elucidate whether altered myokines’ profile is able to improve discriminative potency of contemporary predictive scores.

Acknowledgements

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

REFERENCES

1. Oktay AA, Rich JD, Shah SJ. The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep. 2013; 10(4): 401-10. [DOI: 10.1007/s11897-013-0155-7].

2. Lesyuk W, Kriza C, Kolominsky-Rabas P. Cost-of-illness studies in heart failure: a systematic review 2004-2016. BMC Cardiovasc Disord. 2018; 18(1): 74. [DOI: 10.1186/s12872-018-0815-3].

3. Ziaeian B, Fonarow GC. Epidemiology and aetiology of heart failure. Nat Rev Cardiol. 2016; 13(6): 368-78. [DOI: 10.1038/nrcardio.2016.25].

4. van Riet EE, Hoes AW, Wagenaar KP, Limburg A, Landman MA, Rutten FH. Epidemiology of heart failure: the prevalence of heart failure and ventricular dysfunction in older adults over time. A systematic review. Eur J Heart Fail. 2016; 18(3): 242-52. [DOI: 10.1002/ejhf.483].

5. Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2017; 14(10): 591-602. [DOI: 10.1038/nrcardio.2017.65]. Epub 2017 May 11. PMID: 28492288.

6. Orso F, Fabbri G, Maggioni AP. Epidemiology of Heart Failure. Handb Exp Pharmacol. 2017; 243: 15-33. [DOI: 10.1007/164_2016_74. PMID: 27718059].

7. Chaudhry MA. Heart Failure. Curr Hypertens Rev. 2019; 15(1): 7. [DOI: 10.2174/157340211501190129144451]; [PMID: 30729894].

8. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Colvin MM, Drazner MH, Filippatos GS, Fonarow GC, Givertz MM, Hollenberg SM, Lindenfeld J, Masoudi FA, McBride PE, Peterson PN, Stevenson LW, Westlake C. 2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. Circulation. 2017; 136(6): e137-e161. [DOI: 10.1161/CIR.0000000000000509].

9. Paneroni M, Pasini E, Comini L, Vitacca M, Schena F, Scalvini S, Venturelli M. Skeletal Muscle Myopathy in Heart Failure: the Role of Ejection Fraction. Curr Cardiol Rep. 2018; 20(11): 116. [DOI: 10.1007/s11886-018-1056-x]; PMID: 30259199].

10. Song T, Manoharan P, Millay DP, Koch SE, Rubinstein J, Heiny JA, Sadayappan S. Dilated cardiomyopathy-mediated heart failure induces a unique skeletal muscle myopathy with inflammation. Skelet Muscle. 2019; 9(1): 4. [DOI: 10.1186/s13395-019-0189-y]; [PMID: 30678732]; [PMCID: PMC6345027].

11. Brown DA, Perry JB, Allen ME, Sabbah HN, Stauffer BL, Shaikh SR, Cleland JG, Colucci WS, Butler J, Voors AA, Anker SD, Pitt B, Pieske B, Filippatos G, Greene SJ, Gheorghiade M. Expert consensus document: Mitochondrial function as a therapeutic target in heart failure. Nat Rev Cardiol. 2017; 14(4): 238-250. [DOI: 10.1038/nrcardio.2016.203].

12. Eckel J. Chapter 3 - Skeletal Muscle: A Novel Secretory Organ / In: The Cellular Secretome and Organ Crosstalk. Ed. Juergen Eckel, Academic Press, 2018, Pages 65-90. ISBN 9780128095188, [DOI: 10.1016/C2015-0-06271-2]

13. Pedersen BK. Muscle as a secretory organ. Compr Physiol. 2013; 3(3): 1337-62. [DOI: 10.1002/cphy.c120033].

14. Shen S, Gao R, Bei Y, Li J, Zhang H, Zhou Y, Yao W, Xu D, Zhou F, Jin M, Wei S, Wang K, Xu X, Li Y, Xiao J, Li X. Serum Irisin Predicts Mortality Risk in Acute Heart Failure Patients. Cell Physiol Biochem. 2017; 42(2): 615-622. [DOI: 10.1159/000477867].

15. Silvestrini A, Bruno C, Vergani E, Venuti A, Favuzzi AMR, Guidi F, Nicolotti N, Meucci E, Mordente A, Mancini A. Circulating irisin levels in heart failure with preserved or reduced ejection fraction: A pilot study. PLoS One. 2019; 14(1): e0210320. [DOI: 10.1371/journal.pone.0210320].

16. Tromp J, Khan MAF, Mentz RJ, O’Connor CM, Metra M, Dittrich HC, Ponikowski P, Teerlink JR, Cotter G, Davison B, Cleland JGF, Givertz MM, Bloomfield DM, Van Veldhuisen DJ, Hillege HL, Voors AA, van der Meer P. Biomarker Profiles of Acute Heart Failure Patients With a Mid-Range Ejection Fraction. JACC Heart Fail. 2017; 5(7): 507-517. [DOI: 10.1016/j.jchf.2017.04.007].

17. Miller MS, Vanburen P, Lewinter MM, Lecker SH, Selby DE, Palmer BM, Maughan DW, Ades PA, Toth MJ. Mechanisms underlying skeletal muscle weakness in human heart failure: alterations in single fiber myosin protein content and function. Circ Heart Fail. 2009; 2(6): 700-6. [DOI: 10.1161/CIRCHEARTFAILURE.109.876433]. Epub 2009 Sep 24. PMID: 19919996]; [PMCID: PMC2782533].

18. Piepoli MF, Crisafulli A. Pathophysiology of human heart failure: importance of skeletal muscle myopathy and reflexes. Exp Physiol. 2014; 99(4): 609-15. [DOI: 10.1113/expphysiol.2013.074310].

19. Berezin AE, Berezin AA. Impaired function of fibroblast growth factor 23 / Klotho protein axis in prediabetes and diabetes mellitus: Promising predictor of cardiovascular risk. Diabetes Metab Syndr. 2019; 13(4): 2549-2556. [DOI: 10.1016/j.dsx.2019.07.018].

20. Miyagi M, Kinugasa Y, Sota T, Yamada K, Ishisugi T, Hirai M, Yanagihara K, Haruki N, Matsubara K, Kato M, Yamamoto K. Diaphragm Muscle Dysfunction in Patients With Heart Failure. J Card Fail. 2018; 24(4): 209-216. [DOI: 10.1016/j.cardfail.2017.12.004].

21. Witte KK, Clark AL. Why does chronic heart failure cause breathlessness and fatigue? Prog Cardiovasc Dis. 2007; 49(5): 366-84. [DOI: 10.1016/j.pcad.2006.10.003]; [PMID: 17329182].

22. Lavine KJ, Sierra OL. Skeletal muscle inflammation and atrophy in heart failure. Heart Fail Rev. 2017; 22(2): 179-189. [DOI: 10.1007/s10741-016-9593-0].

23. Fülster S, Tacke M, Sandek A, Ebner N, Tschöpe C, Doehner W, Anker SD, von Haehling S. Muscle wasting in patients with chronic heart failure: results from the studies investigating co-morbidities aggravating heart failure (SICA-HF). Eur Heart J. 2013; 34(7): 512-9. [DOI: 10.1093/eurheartj/ehs381].

24. Hamazaki N, Kamiya K, Matsuzawa R, Nozaki K, Ichikawa T, Tanaka S, Nakamura T, Yamashita M, Maekawa E, Noda C, Yamaoka-Tojo M, Matsunaga A, Masuda T, Ako J. Prevalence and prognosis of respiratory muscle weakness in heart failure patients with preserved ejection fraction. Respir Med. 2020; 161: 105834. [DOI: 10.1016/j.rmed.2019.105834.]

25. Chapleau MW. Contributions of skeletal muscle myopathy to heart failure: novel mechanisms and therapies. Introduction. Exp Physiol. 2014; 99(4): 607-8. [DOI: 10.1113/expphysiol.2013.074294].

26. Kalkan AK, Cakmak HA, Erturk M, Kalkan KE, Uzun F, Tasbulak O, Diker VO, Aydin S, Celik A. Adropin and Irisin in Patients with Cardiac Cachexia. Arq Bras Cardiol. 2018; 111(1): 39-47. [DOI: 10.5935/abc.20180109.

27. Piccirillo R. Exercise-Induced Myokines With Therapeutic Potential for Muscle Wasting. Front Physiol. 2019; 10: 287. [DOI: 10.3389/fphys.2019.00287].

28. Brum PC, Bacurau AV, Cunha TF, Bechara LR, Moreira JB. Skeletal myopathy in heart failure: effects of aerobic exercise training. Exp Physiol. 2014; 99(4): 616-20. [DOI: 10.1113/expphysiol.2013.076844].

29. Vu TT, Marquez J, Le LT, Nguyen ATT, Kim HK, Han J. The role of decorin in cardiovascular diseases: more than just a decoration. Free Radic Res. 2018; 52(11-12): 1210-1219. [DOI: 10.1080/10715762.2018.1516285.

30. Jahanyar J, Joyce DL, Southard RE, Loebe M, Noon GP, Koerner MM, Torre-Amione G, Youker KA. Decorin-mediated transforming growth factor-beta inhibition ameliorates adverse cardiac remodeling. J Heart Lung Transplant. 2007; 26(1): 34-40. [DOI: 10.1016/j.healun.2006.10.005].

31. Colaianni G, Cinti S, Colucci S, Grano M. Irisin and musculoskeletal health. Ann N Y Acad Sci. 2017; 1402(1): 5-9. [DOI: 10.1111/nyas.13345].

32. Abd El-Mottaleb NA, Galal HM, El Maghraby KM, Gadallah AI. Serum irisin level in myocardial infarction patients with or without heart failure. Can J Physiol Pharmacol. 2019; 97(10): 932-938. [DOI: 10.1139/cjpp-2018-0736.

33. Seldin MM, Peterson JM, Byerly MS, Wei Z, Wong GW. Myonectin (CTRP15), a novel myokine that links skeletal muscle to systemic lipid homeostasis. J Biol Chem. 2012; 287(15): 11968-80. [DOI: 10.1074/jbc.M111.336834].

34. Otaka N, Shibata R, Ohashi K, Uemura Y, Kambara T, Enomoto T, Ogawa H, Ito M, Kawanishi H, Maruyama S, Joki Y, Fujikawa Y, Narita S, Unno K, Kawamoto Y, Murate T, Murohara T, Ouchi N. Myonectin Is an Exercise-Induced Myokine That Protects the Heart From Ischemia-Reperfusion Injury. Circ Res. 2018; 123(12): 1326-1338. [DOI: 10.1161/CIRCRESAHA.118.313777].

35. McKay BR, Nederveen JP, Fortino SA, Snijders T, Joanisse S, Kumbhare DA, Parise G. Brain-derived neurotrophic factor is associated with human muscle satellite cell differentiation in response to muscle-damaging exercise. Appl Physiol Nutr Metab. 2020; 45(6): 581-590. [DOI: 10.1139/apnm-2019-0501].

36. Petyunina O, Kopytsya M, Berezin A. Brain-derived neurotrophic factor gene polymorphism in post-ST-elevation myocardial infarction patients undergoing primary percutaneous intervention. Biomedical Research and Therapy. 2020; 7(8): 3921-3932. [DOI: 10.15419/bmrat.v7i8.622]

37. Yeghiazarians Y, Honbo N, Imhof I, Woods B, Aguilera V, Ye J, Boyle AJ, Karliner JS. IL-15: a novel prosurvival signaling pathway in cardiomyocytes. J Cardiovasc Pharmacol. 2014; 63(5): 406-11. [DOI: 10.1097/FJC.0000000000000061]; [PMID: 24805144]; [PMCID: PMC4115639].

38. Baán JA, Varga ZV, Leszek P, Kuśmierczyk M, Baranyai T, Dux L, Ferdinandy P, Braun T, Mendler L. Myostatin and IGF-I signaling in end-stage human heart failure: a qRT-PCR study. J Transl Med. 2015; 13: 1. [DOI: 10.1186/s12967-014-0365-0].

39. George I, Bish LT, Kamalakkannan G, Petrilli CM, Oz MC, Naka Y, Sweeney HL, Maybaum S. Myostatin activation in patients with advanced heart failure and after mechanical unloading. Eur J Heart Fail. 2010; 12(5): 444-53. [DOI: 10.1093/eurjhf/hfq039].

40. Nielsen AR, Pedersen BK. The biological roles of exercise-induced cytokines: IL-6, IL-8, and IL-15. Appl Physiol Nutr Metab. 2007; 32(5): 833-9. [DOI: 10.1139/H07-054].

41. Ueland T, Gullestad L, Nymo SH, Yndestad A, Aukrust P, Askevold ET. Inflammatory cytokines as biomarkers in heart failure. Clin Chim Acta. 2015; 443: 71-7. [DOI: 10.1016/j.cca.2014.09.001]

42. Berezin A.. Bone-Related Proteins as Markers in Vascular Remodeling. In: Preedy V. (eds) Biomarkers in Bone Disease. Biomarkers in Disease: Methods, Discoveries and Applications. Springer, Dordrecht. 2015, pp. 1-22. [DOI: 10.1007/978-94-007-7745-3_4-1]

43. Shen S, Gao R, Bei Y, Li J, Zhang H, Zhou Y, Yao W, Xu D, Zhou F, Jin M, Wei S, Wang K, Xu X, Li Y, Xiao J, Li X. Serum Irisin Predicts Mortality Risk in Acute Heart Failure Patients. Cell Physiol Biochem. 2017; 42(2): 615-622. [DOI: 10.1159/000477867].

44. Kalkan AK, Cakmak HA, Erturk M, Kalkan KE, Uzun F, Tasbulak O, Diker VO, Aydin S, Celik A. Adropin and Irisin in Patients with Cardiac Cachexia. Arq Bras Cardiol. 2018; 111(1): 39-47. [DOI: 10.5935/abc.20180109]

45. Chen P, Liu Z, Luo Y, Chen L, Li S, Pan Y, Lei X, Wu D, Xu D. Predictive value of serum myostatin for the severity and clinical outcome of heart failure. Eur J Intern Med. 2019; 64: 33-40. [DOI: 10.1016/j.ejim.2019.04.017].

46. Barman HA, Şahin I, Atıcı A, Durmaz E, Yurtseven E, Ikitimur B, Okuyan E, Keleş I. Prognostic significance of brain-derived neurotrophic factor levels in patients with heart failure and reduced left ventricular ejection fraction. Anatol J Cardiol. 2019; 22(6): 309-316. [DOI: 10.14744/AnatolJCardiol.2019.37941].

47. Berezin AE, Kremzer AA. Predictive value of circulating osteonectin in patients with ischemic symptomatic chronic heart failure. Biomed J. 2015; 38(6): 523-30. [DOI: 10.1016/j.bj.2015.08.002].

Refbacks

  • There are currently no refbacks.


Creative Commons License
This work is licensed under a Creative Commons Attribution 3.0 License.