Journal Home > Volume 18 , Issue 1

Heart failure is common in adult population, accounting for substantial morbidity and mortality worldwide. The main risk factors for heart failure are coronary artery disease, hypertension, obesity, diabetes mellitus, chronic pulmonary diseases, family history of cardiovascular diseases, cardiotoxic therapy. The main factor associated with poor outcome of these patients is constant progression of heart failure. In the current review we present evidence on the role of established and candidate neurohumoral biomarkers for heart failure progression management and diagnostics. A growing number of biomarkers have been proposed as potentially useful in heart failure patients, but not one of them still resembles the characteristics of the “ideal biomarker.” A single marker will hardly perform well for screening, diagnostic, prognostic, and therapeutic management purposes. Moreover, the pathophysiological and clinical significance of biomarkers may depend on the presentation, stage, and severity of the disease. The authors cover main classification of heart failure phenotypes, based on the measurement of left ventricular ejection fraction, including heart failure with preserved ejection fraction, heart failure with reduced ejection fraction, and the recently proposed category heart failure with mid-range ejection fraction. One could envisage specific sets of biomarker with different performances in heart failure progression with different left ventricular ejection fraction especially as concerns prediction of the future course of the disease and of left ventricular adverse/reverse remodeling. This article is intended to provide an overview of basic and additional mechanisms of heart failure progression will contribute to a more comprehensive knowledge of the disease pathogenesis.


menu
Abstract
Full text
Outline
About this article

Neurohumoral, cardiac and inflammatory markers in the evaluation of heart failure severity and progression

Show Author's information Ekaterina A Polyakova( )Evgeny N MikhaylovDmitry L SoninYuri V CheburkinMikhail M Galagudza
Almazov National Medical Research Centre, Saint-Petersburg, Russia

Abstract

Heart failure is common in adult population, accounting for substantial morbidity and mortality worldwide. The main risk factors for heart failure are coronary artery disease, hypertension, obesity, diabetes mellitus, chronic pulmonary diseases, family history of cardiovascular diseases, cardiotoxic therapy. The main factor associated with poor outcome of these patients is constant progression of heart failure. In the current review we present evidence on the role of established and candidate neurohumoral biomarkers for heart failure progression management and diagnostics. A growing number of biomarkers have been proposed as potentially useful in heart failure patients, but not one of them still resembles the characteristics of the “ideal biomarker.” A single marker will hardly perform well for screening, diagnostic, prognostic, and therapeutic management purposes. Moreover, the pathophysiological and clinical significance of biomarkers may depend on the presentation, stage, and severity of the disease. The authors cover main classification of heart failure phenotypes, based on the measurement of left ventricular ejection fraction, including heart failure with preserved ejection fraction, heart failure with reduced ejection fraction, and the recently proposed category heart failure with mid-range ejection fraction. One could envisage specific sets of biomarker with different performances in heart failure progression with different left ventricular ejection fraction especially as concerns prediction of the future course of the disease and of left ventricular adverse/reverse remodeling. This article is intended to provide an overview of basic and additional mechanisms of heart failure progression will contribute to a more comprehensive knowledge of the disease pathogenesis.

References(160)

[1]

Wintrich J, Kindermann I, Böhm M. Neues zur Herzinsuffizienz [Update on heart failure]. Herz 2020; 45: 158−169.

[2]

Mosterd A, Hoes AW. Clinical epidemiology of heart failure. Heart 2007; 93: 1137−1146.

[3]

Arfsten H, Bartko P E, Pavo N, et al. Phenotyping progression of secondary mitral regurgitation in chronic systolic heart failure. Eur J Clin Invest 2019; 49: e13159.

[4]

Cuthbert JJ, Pellicori P, Clark AL. Cardiovascular outcomes with sacubitril-valsartan in heart failure: emerging clinical data. Ther Clin Risk Manag 2020; 16: 715−726.

[5]

Nakata T, Hashimoto A, Moroi M, et al. Sudden death prediction by C-reactive protein, electrocardiographic findings, and myocardial fatty acid uptake in haemodialysis patients: analysis of a multicentre prospective cohort sub-study. Eur Heart J Cardiovasc Imaging 2016; 17: 1394−1404.

[6]

Buckley LF, Shah AM. Recent advances in the treatment of chronic heart failure [version, 1,peer review: 3 approved]. F1000Research 2019; 8(F1000 Faculty Rev): 2134.

[7]

Piek A, Du W, de Boer RA, Silljé, H HW. Novel heart failure biomarkers: why do we fail to exploit their potential? Crit Rev Clin Lab Sci 2018; 55: 246−263.

[8]

Carnes J, Gordon G. Biomarkers in heart failure with preserved ejection fraction: an update on progress and future challenges. Heart Lung Circ 2020; 29: 62−68.

[9]

Peirlinck M, Sahli Costabal F, Sack KL, et al. Using machine learning to characterize heart failure across the scales. Biomech Model Mechanobiol 2019; 18: 1987−2001.

[10]

Jorba G, Aguirre-Plans J, Junet V, et al. In-silico simulated prototype-patients using TPMS technology to study a potential adverse effect of sacubitril and valsartan. PLoS One 2020; 15: e0228926.

[11]

Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J 2016; 37: 2129−2200.

[12]

Adachi M, Watanabe M, Kurata Y, et al. β-Adrenergic Blocker, Carvedilol, Abolishes Ameliorating Actions of Adipose-Derived Stem Cell Sheets on Cardiac Dysfunction and Remodeling After Myocardial Infarction. Circ J 2019; 83: 2282−2291.

[13]

Senni M, Wachter R, Witte KK, et al. Initiation of sacubitril/valsartan shortly after hospitalisation for acutely decompensated heart failure in patients with newly diagnosed (de novo) heart failure: a subgroup analysis of the TRANSITION study. Eur J Heart Fail 2019; 22: 303−312.

[14]

Koba S, Hisatome I, Watanabe T. Augmented fear bradycardia in rats with heart failure. J Physiol Sci 2019; 69: 875−883.

[15]

Redeker NS, Conley S, Anderson G, et al. Effects of cognitive behavioral therapy for insomnia on sleep, symptoms, stress, and autonomic function among patients with heart failure. Behav Sleep Med 2020; 18: 190−202.

[16]

Florea VG, Rector TS, Anand IS, Cohn JN. Heart failure with improved ejection fraction: clinical characteristics, correlates of recovery, and survival: results from the valsartan heart failure trial. Circ Heart Fail 2016; 9: e003123.

[17]

Tarantino N, Santoro F Di Biase L, et al. Chromogranin-A serum levels in patients with takotsubo syndrome and ST elevation acute myocardial infarction. Int J Cardiol 2020; 320: 12−17.

[18]

Guellich A, Mehel H, Fischmeister R. Cyclic AMP synthesis and hydrolysis in the normal and failing heart. Pflugers Arch 2014; 466: 1163−1175.

[19]

Belmonte SL, Blaxall BC. G protein coupled receptor kinases as therapeutic targets in cardiovascular disease. Circ Res 2011; 109: 309−319.

[20]

Habecker BA, Anderson ME, Birren SJ, et al. Molecular and cellular neurocardiology: development, and cellular and molecular adaptations to heart disease. J Physiol 2016; 594: 3853−3875.

[21]

Borovac JA, D'Amario D, Bozic J, Glavas D. Sympathetic nervous system activation and heart failure: Current state of evidence and the pathophysiology in the light of novel biomarkers. World J Cardiol 2020; 12: 373−408.

[22]

Maisel AS, Scott NA, Motulsky HJ, et al. Elevation of plasma neuropeptide Y levels in congestive heart failure. Am J Med 1989; 86: 43−48.

[23]

Ajijola OA, Chatterjee NA, Gonzales MJ, et al. Coronary Sinus Neuropeptide Y Levels and Adverse Outcomes in Patients With Stable Chronic Heart Failure. JAMA Cardiol 2020; 5: 318−325.

[24]

Mahata SK, Mahapatra NR, Mahata M, et al. Catecholamine secretory vesicle stimulus-transcription coupling in vivo. J Biol Chem 2003; 278: 32058−67.

[25]

Penna C, Alloatti G, Gallo MP, et al. Catestatin improves post-ischemic left ventricular function and decreases ischemia/reperfusion injury in heart. Cell Mol Neurobiol 2010; 30: 1171−1179.

[26]

Zhu D, Wang F, Yu H, et al. Catestatin is useful in detecting patients with stage B heart failure. Biomarkers 2011; 16: 691−697.

[27]

Hodatsu A, Fujino N, Uyama Y et al. Impact of cardiac myosin light chain kinase gene mutation on development of dilated cardiomyopathy. ESC Heart Fail 2019; 6: 406−415.

[28]

Roffe-Vazquez DN, Huerta-Delgado AS, Castillo EC, et al. Correlation of Vitamin D with Inflammatory Cytokines, Atherosclerotic Parameters, and Lifestyle Factors in the Setting of Heart Failure: A 12-Month Follow-Up Study. Int J Mol Sci 2019; 20: 5811.

[29]

Lubrano V, Balzan S. Role of oxidative stress-related biomarkers in heart failure: galectin 3, α1-antitrypsin and LOX-1: new therapeutic perspective? Mol Cell Biochem 2019; 464: 143−152.

[30]

Jia X, Sun W, Hoogeveen RC, et al. Response by Jia, et al to Letter Regarding Article, “High-Sensitivity Troponin I and Incident Coronary Events, Stroke, Heart Failure Hospitalization, and Mortality in the ARIC Study.”. Circulation 2019; 140: e772−e773.

[31]

Islam MN, Chowdhury MS, Paul GK, et al. Association of Diastolic Dysfunction with N-terminal Pro-B-type Natriuretic Peptide Level in Heart Failure Patients with Preserved Ejection Fraction. Mymensingh Med J 2019; 28: 333−346.

[32]

Motiwala SR, Januzzi JL. The role of natriuretic peptides as biomarkers for guiding the management of chronic heart failure. Clin Pharmacol Ther 2013; 93: 57−67.

[33]

Vanderheyden M, Bartunek J, Goethals M, et al. Dipeptidyl-peptidase IV and B-type natriuretic peptide. From bench to bedside. Clin Chem Lab Med 2009; 47: 248−252.

[34]
Djordjevic T, Arena R, Guazzi M, Popovic D. Prognostic Value of NT-Pro Brain Natriuretic Peptide During Exercise Recovery in Ischemic Heart Failure of Reduced, Midrange, and Preserved Ejection Fraction. J Cardiopulm Rehabil Prev. 2020; Published online first: Sep 16 2020. Doi: 10.1097/HCR.0000000000000531.
[35]

Salah K, Stienen S, Pinto YM, et al. Prognosis and NT-proBNP in heart failure patients with preserved versus reduced ejection fraction. Heart 2019; 105: 1182−1189.

[36]

Januzzi JL Jr, Camargo CA, Anwaruddin S, et al. The N-terminal Pro-BNP investigation of dyspnea in the emergency department (PRIDE) study. Am J Cardiol 2005; 95: 948−954.

[37]

Simpson J, Castagno D, Doughty RN, et al. Is heart rate a risk marker in patients with chronic heart failure and concomitant atrial fibrillation? Results from the MAGGIC meta-analysis. Eur J Heart Fail 2015; 17: 1182−1191.

[38]

Moertl D, Berger R, Struck J, et al. Comparison of midregional pro-atrial and B-type natriuretic peptides in chronic heart failure: influencing factors, detection of left ventricular systolic dysfunction, and prediction of death. J Am Coll Cardiol 2009; 53: 1783−1790.

[39]

Emdin M, Vittorini S, Passino C, Clerico A. Old and new biomarkers of heart failure. Eur J Heart Fail 2009; 11: 331−335.

[40]

Huang YT, Tseng YT, Chu TW, et al. N-terminal pro b-type natriuretic peptide (NT-pro-BNP) –based score can predict in-hospital mortality in patients with heart failure. Scientific Reports 2016; 6: 32902.

[41]

Kociol RD, Horton JR, Fonarow GC, et al. Admission, discharge, or change in B-type natriuretic peptide and long-term outcomes: data from Organized Program to Initiate Lifesaving Treatment in Hospitalized Patients with Heart Failure (OPTIMIZE-HF) linked to Medicare claims. Circ Heart Fail 2011; 4: 628−636.

[42]

de Bold AJ, Borenstein HB, Veress AT, Sonnenberg H. A rapid and potent natriuretic response to intravenous injection of atrial myocardial extract in rats. Life Sci 1981; 28: 89−94.

[43]

Sanz MP, Borque L, Rus A, et al. Comparison of BNP and NT-proBNP assays in the approach to the emergency diagnosis of acute dyspnea. J Clin Lab Analysis 2006; 20: 227−232.

[44]

Maisel A, Mueller C, Nowak RM, et al. Midregion prohormone adrenomedullin and prognosis in patients presenting with acute dyspnea: results from the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll Cardiol 2011; 58: 1057−1067.

[45]

Möckel M, von Haehling S, Vollert JO, et al. BACH study group. Early identification of acute heart failure at the time of presentation: do natriuretic peptides make the difference? ESC Heart Fail 2018; 5: 309−315.

[46]

Abdul-Rahim AH, Perez AC, Fulton RL, et al. Risk of stroke in chronic heart failure patients without atrial fibrillation: analysis of the controlled rosuvastatin in multinational trial heart failure (CORONA) and the Gruppo Italiano per lo Studio della Sopravvivenza nell’Insufficienza Cardiaca-Heart Failure (GISSI-HF) trials. Circulation 2015; 131: 1486−1494.

[47]

Gierula J, Cubbon RM, Paton MF, et al. Prospective evaluation and long term follow up of patients referred to secondary care based upon natriuretic peptide levels in primary care. Eur Heart J Qual Care Clin Outcomes 2019; 5: 218−224.

[48]

Kjeldsen SE, von Lueder TG, Smiseth OA, et al. Medical Therapies for Heart Failure With Preserved Ejection Fraction. Hypertension 2000; 75: 23−32.

[49]

Arakawa HUaK. Angiotensin II-forming systems in cardiovascular dis- eases. Heart Fail Rev 1998; 3: 119−124.

[50]

Dendorfer A, Thornagel A, Raasch W, et al. Angiotensin II induces catecholamine release by direct ganglionic excitation. Hypertension 2002; 40: 348−354.

[51]

Weber KT, Brilla CG. Pathological hypertrophy and cardiac interstitium. Fibrosis and renin-angiotensin-aldosterone system. Circulation 1991; 83: 1849−1865.

[52]

Sullivan RD, Mehta RM, Tripathi R, et al. Renin Activity in Heart Failure with Reduced Systolic Function—New Insights. Int J Mol Sci 2019; 20: 3182.

[53]

Vergaro G, Emdin M, Iervasi A, et al. Prognostic value of plasma renin activity in heart failure. Am J Cardiol 2011; 108: 246−251.

[54]

Bhandari SK, Batech M, Shi J, et al. Plasma renin activity and risk of cardiovascular and mortality outcomes among individuals with elevated and nonelevated blood pressure. Kidney Res. Clin Pract 2016; 35: 219−228.

[55]

Latini R, Masson S, Anand I, et al. The comparative prognostic value of plasma neurohormones at baseline in patients with heart failure enrolled in Val-HeFT. Eur Heart J 2004; 25: 292−299.

[56]

Tsutamoto T, Sakai H, Tanaka T. Comparison of active renin concentration and plasma renin activity as a prognostic predictor in patients with heart failure. Circ J 2007; 71: 915−921.

[57]

Kavsak PA, Tandon V, Ainsworth C. A Three-Site Immunoassay for High-Sensitivity Cardiac Troponin I with Low Immunoreactivity for Macrocomplexes. Clin Chem 2020; 66: 854−855.

[58]

Masotti S, Prontera C, Musetti V, et al. Evaluation of analytical performance of a new high-sensitivity immunoassay for cardiac troponin I. Clin Chem Lab Med 2018; 56: 492−501.

[59]

Park KC, Gaze DC, Collinson PO, Marber MS. Cardiac troponins: from myocardial infarction to chronic disease. Cardiovasc Res 2017; 113: 1708−1718.

[60]

Weil BR, Suzuki G, Young RF. Troponin Release and Reversible Left Ventricular Dysfunction After Transient Pressure Overload. J Am Coll Cardiol 2018; 71: 2906−2916.

[61]

de Lemos JA, Drazner MH, Omland T, et al. Association of troponin T detected with a highly sensitive assay and cardiac structure and mortality risk in the general population. JAMA 2010; 304: 2503−2512.

[62]

Oluleye OW, Folsom AR, Nambi V, et al. Troponin T, B-type natriuretic peptide, C-reactive protein, and cause-specific mortality. Ann Epidemiol 2013; 23: 66−73.

[63]

Evans JDW, Dobbin SJH, Pettit SJ, et al. High-Sensitivity Cardiac Troponin and New-Onset Heart Failure: A Systematic Review and Meta-Analysis of 67,063 Patients With 4,165 Incident Heart Failure Events. JACC Heart Fail 2018; 6: 187−197.

[64]

Nagarajan V, Hernandez AV, Tang WH. Prognostic value of cardiac troponin in chronic stable heart failure: a systematic review. Heart 2012; 98: 1778−1786.

[65]

Latini R, Masson S, Inder SA, et al. Prognostic value of very low plasma concentrations of troponin T in patients with stable chronic heart failure. Circulation 2007; 116: 1242−1249.

[66]

Kim KK, Sheppard D, Chapman HA. TGF-β1 Signaling and Tissue Fibrosis. Cold Spring Harb Perspect Biol 2018; 10: a022293.

[67]

Purnomo Y, Piccart Y, Coenen T, et al. Oxidative stress and transforming growth factor-β1-induced cardiac fibrosis. Cardiovasc Hematol Disord Drug Targets 2013; 13: 165−172.

[68]
Schultz GS, Chin GA, Moldawer L, et al. Principles of Wound Healing. In: Fitridge R, Thompson M, editors. Mechanisms of Vascular Disease: A Reference Book for Vascular Specialists. Adelaide (AU): University of Adelaide Press, 2011.
[69]

Hinz B. The extracellular matrix and transforming growth factor-β1: Tale of a strained relationship. Matrix Biol 2015; 47: 54−65.

[70]

Liu RM, Desai LP. Reciprocal regulation of TGF-β and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol 2015; 6: 565−577.

[71]

Humeres C, Frangogiannis NG. Fibroblasts in the Infarcted, Remodeling, and Failing Heart. JACC Basic Transl Sci 2019; 4: 449−467.

[72]

Akhurst RJ. Targeting TGF-β Signaling for Therapeutic Gain. Cold Spring Harb Perspect Biol 2017; 9: a022301.

[73]

de Boer RA, Yu L, van Veldhuisen DJ. Galectin-3 in cardiac remodeling and heart failure. Curr Heart Fail Rep 2010; 7: 1−8.

[74]

Zhong X, Qian X, Chen G, Song X. The role of galectin-3 in heart failure and cardiovascular disease. Clin Exp Pharmacol Physiol 2019; 46: 197−203.

[75]

Henderson NC, Sethi T. The regulation of inflammation by galectin-3. Immunol Rev 2009; 230: 160−171.

[76]

Vergaro G, Prud'homme M, Fazal L, et al. Inhibition of Galectin-3 Pathway Prevents Isoproterenol-Induced Left Ventricular Dysfunction and Fibrosis in Mice. Hypertension 2016; 67: 606−612.

[77]

van Kimmenade RR, Januzzi JL Jr, Ellinor PT, et al. Utility of amino-terminal pro-brain natriuretic peptide, galectin-3, and apelin for the evaluation of patients with acute heart failure. J Am Coll Cardiol 2006; 48: 1217−1224.

[78]

Shah AM, Mann DL. In search of new therapeutic targets and strategies for heart failure: recent advances in basic science. Lancet 2011; 378: 704−712.

[79]

Lok DJ, Van Der Meer P, de la Porte PW, et al. Prognostic value of galectin-3, a novel marker of fibrosis, in patients with chronic heart failure: data from the DEAL-HF study. Clin Res Cardiol 2010; 99: 323−328.

[80]

Felker GM, Fiuzat M, Shaw LK, et al. Galectin-3 in ambulatory patients with heart failure: results from the HF-ACTION study. Circ Heart Fail 2012; 5: 72−78.

[81]

Gullestad L, Ueland T, Kjekshus J, et al. Galectin-3 predicts response to statin therapy in the Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA). Eur Heart J 2012; 33: 2290−2296.

[82]

Gandhi PU, Motiwala SR, Belcher AM, et al. Galectin-3 and mineralocorticoid receptor antagonist use in patients with chronic heart failure due to left ventricular systolic dysfunction. Am Heart J 2015; 169: 404−411.

[83]

Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2013; 62: e147−e239.

[84]

Carrasco-Sánchez FJ, Aramburu-Bodas O, Salamanca-Bautista P, et al. Predictive value of serum galectin-3 levels in patients with acute heart failure with preserved ejection fraction. Int J Cardiol 2013; 169: 177−182.

[85]

Edelmann F, Holzendorf V, Wachter R, et al. Galectin-3 in patients with heart failure with preserved ejection fraction: results from the Aldo-DHF trial. Eur J Heart Fail 2015; 17: 214−223.

[86]

Wu C-K, Su M-Y, Lee J-K, et al. Galectin-3 level and the severity of cardiac diastolic dysfunction using cellular and animal models and clinical indices. Sci Rep 2015; 5: 17007.

[87]

González GE, Cassaglia P, Noli Truant S, et al. Galectin-3 is essential for early wound healing and ventricular remodeling after myocardial infarction in mice. Int J Cardiol 2014; 176: 1423−1425.

[88]

de Boer RA, De Keulenaer G, Bauersachs J, et al. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur J Heart Fail 2019; 21: 272−285.

[89]

Pascual-Figal DA, Januzzi JL. The biology of ST2: International ST2 Consensus Panel. Am J Cardiol 2015; 115: 3B−7B.

[90]

Sanada S, Hakuno D, Higgins LJ, et al. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest 2007; 117: 1538−1549.

[91]

Shah RV, Chen-Tournoux AA, Picard MH, et al. Serum levels of the interleukin-1 receptor family member ST2, cardiac structure and function, and long-term mortality in patients with acute dyspnea. Circ Heart Fail 2009; 2: 311−319.

[92]

Dieplinger B, Januzzi JL Jr, Steinmair M, et al. Analytical and clinical evaluation of a novel high-sensitivity assay for measurement of soluble ST2 in human plasma--the Presage ST2 assay. Clin Chim Acta 2009; 409: 33−40.

[93]

Shah KB, Kop WJ, Christenson RH, et al. Prognostic utility of ST2 in patients with acute dyspnea and preserved left ventricular ejection fraction. Clin Chem 2011; 57: 874−882.

[94]

Manzano-Fernández S, Mueller T, Pascual-Figal D, et al. Usefulness of soluble concentrations of interleukin family member ST2 as predictor of mortality in patients with acutely decompensated heart failure relative to left ventricular ejection fraction. Am J Cardiol 2011; 107: 259−267.

[95]

Januzzi JL Jr, Peacock WF, Maisel AS, et al. Measurement of the interleukin family member ST2 in patients with acute dyspnea: results from the PRIDE (Pro-Brain Natriuretic Peptide Investigation of Dyspnea in the Emer- gency Department) study. J Am Coll Cardiol 2007; 50: 607−613.

[96]

Rehman SU, Mueller T, Januzzi JL Jr. Characteristics of the novel interleukin family biomarker ST2 in patients with acute heart failure. J Am Coll Cardiol 2008; 52: 1458−1465.

[97]

Anand IS, Rector TS, Kuskowski M, et al. Prognostic value of soluble ST2 in the Valsartan Heart Failure Trial. Circ Heart Fail 2014; 7: 418−426.

[98]

Villacorta H, Maisel AS. Soluble ST2 Testing: A Promising Biomarker in the Management of Heart Failure. Arq Bras Cardiol 2016; 106: 145−152.

[99]

McMurray JJ, Kjekshus J, Gullestad L, et al. Effects of statin therapy according to plasma high- sensitivity C-reactive protein concentration in the Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA): a retrospective analysis. Circulation 2009; 120: 2188−2196.

[100]

Gaggin HK, Szymonifka J, Bhardwaj A, et al. Head-to-head comparison of serial soluble ST2, growth differentiation factor-15, and highly-sensitive troponin T measurements in patients with chronic heart failure. JACC Heart Fail 2014; 2: 65−72.

[101]

Groenewegen A, Rutten FH, Mosterd A, Hoes AW. Epidemiology of heart failure. Eur J Heart Fail 2020; 22: 1342−1356.

[102]

Kuhl M, Sheldahl LC, Park M, et al. The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet 2000; 16: 279−283.

[103]

Anand IS, Latini R, Florea VG, et al. C-reactive protein in heart failure: prognostic value and the effect of valsartan. Circulation 2005; 112: 1428−1434.

[104]

Sola S, Mir MQ, Lerakis S, et al. Atorvastatin improves left ventricular systolic function and serum markers of inflammation in nonischemic heart failure. J Am Coll Cardiol 2006; 47: 332−337.

[105]

Mann DL, McMurray JJ, Packer M, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation 2004; 109: 1594−1602.

[106]

Chung ES, Packer M, Lo KH, et al. Anti-TNF Therapy Against Congestive Heart Failure Investigators. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 2003; 107: 3133−3140.

[107]

Düring J, Annborn M, Cronberg T, et al. Copeptin as a marker of outcome after cardiac arrest: a sub-study of the TTM trial. Crit Care 2020; 24: 185.

[108]

Maisel A, Xue Y, Shah K. Increased 90-day mortality in patients with acute heart failure with elevated copeptin: secondary results from the Biomarkers in Acute Heart Failure (BACH) study. Circ Heart Fail 2011; 4: 613−620.

[109]

Pozsonyi Z, Förhécz Z, Gombos T, et al. Copeptin (C-terminal pro arginine-vasopressin) is an independent long-term prognostic marker in heart failure with reduced ejection fraction. Heart Lung Circ 2015; 24: 359−367.

[110]

Voors AA, Kremer D, Geven C, et al. Adrenomedullin in heart failure: pathophysiology and therapeutic application. Eur J Heart Fail 2019; 21: 163−171.

[111]

Shah RV, Truong QA, Gaggin HK, et al. Mid-regional pro-atrial natriuretic peptide and pro-adrenomedullin testing for the diagnostic and prognostic evaluation of patients with acute dyspnoea. Eur Heart J 2012; 33: 2197−2205.

[112]

Tarzami ST. Chemokines and inflammation in heart disease: Adaptive or maladaptive? Int J Clin Exp Med 2011; 4: 74−80.

[113]

Zhang D, Fan GC, Zhou X, et al. Over-expression of CXCR4 on mesenchymal stem cells augments myoangiogenesis in the infarcted myocardium. J Mol Cell Cardiol 2019; 44: 281−292.

[114]

Domouzoglou EM, Naka KK, Vlahos AP, et al. Fibroblast growth factors in cardiovascular disease: The emerging role of FGF21. Am J Physiol Heart Circ Physiol 2015; 309: H1029−H1038.

[115]

Planavila A, Fernández-Solà J, Villarroya F. Cardiokines as Modulators of Stress-Induced Cardiac Disorders. Adv Protein Chem Struct Biol 2017; 108: 227−256.

[116]

Karmazyn M, Purdham DM, Rajapurohitam V, Zeidan A. Signalling mechanisms underlying the metabolic and other effects of adipokines on the heart. Cardiovasc Res 2008; 79: 279−286.

[117]

Schram K, Sweeney G. Implications of myocardial matrix remodeling by adipokines in obesity-related heart failure. Trends Cardiovasc Med 2008; 18: 199−205.

[118]

Du Y, Wang X, Li L, et al. miRNA-Mediated Suppression of a Cardioprotective Cardiokine as a Novel Mechanism Exacerbating Post-MI Remodeling by Sleep Breathing Disorders. Circ Res 2020; 126: 212−228.

[119]

Landecho MF, Tuero C, Valentí V, et al. Relevance of Leptin and Other Adipokines in Obesity-Associated Cardiovascular Risk. Nutrients 2019; 11: 2664.

[120]

Liu Y, Vu V, Sweeney G. Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases. Front Endocrinol (Lausanne) 2019; 10: 842.

[121]

Peri-Okonny PA, Ayers C, Maalouf N, et al. Adiponectin protects against incident hypertension independent of body fat distribution: observations from the Dallas Heart Study. Diabetes Metab Res Rev 2017; 33.

[122]

Ebrahimi-Mamaeghani M, Mohammadi S, Arefhosseini SR, et al. Adiponectin as a potential biomarker of vascular disease. Vasc Health Risk Manag 2015; 11: 55−70.

[123]

Achari AE, Jain SK. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci 2017; 18: 1321.

[124]

Gruzdeva OV, Borodkina AD, Akbasheva OE, et al. Influence of visceral obesity on the secretion of adipokines with epicardial adipocytes in patients with coronary heart disease. Ter Arkh 2018; 90: 71−78.

[125]

Wong C, Marwick TH. Obesity cardiomyopathy: pathogenesis and pathophysiology. Nat Clin Pract Cardiovasc Med 2007; 4: 436−443.

[126]

Smith CCT, Yellon DM. Adipocytokines, cardiovascular pathophysiology and myocardial protection. Pharmacol Ther 2011; 129: 206−219.

[127]

Symons JD, Abel ED. Lipotoxicity contributes to endothelial dysfunction: a focus on the contribution from ceramide. Rev Endocr Metab Disord 2013; 14: 59−68.

[128]

Li P, Shibata R, Unno K, et al. Evidence for the importance of adiponectin in the cardioprotective effects of pioglitazone. Hypertension 2010; 55: 69−75.

[129]

Hui X, Lam KS, Vanhoutte PM, Xu A. Adiponectin and cardiovascular health: an update. Br J Pharmacol 2012; 165: 574−590.

[130]

Smith CC, Mocanu MM, Davidson SM, et al. Leptin, the obesity-associated hormone, exhibits direct cardioprotective effects. Br J Pharmacol 2006; 149: 5−13.

[131]

Chubenko EA, Belyaeva OD, Berkovich OA, Baranova EI. Leptin and metabolic syndrome. Russian Journal of Physiology (formerly I. M. Sechenov Physiological Journal) 2010; 96: N10.

[132]

Poetsch MS, Strano A, Guan K. Role of Leptin in Cardiovascular Diseases. Front Endocrinol (Lausanne) 2020; 11: 354.

[133]

Bairwa SC, Rajapurohitam V, Gan XT, et al. Cardiomyocyte Antihypertrophic Effect of Adipose Tissue Conditioned Medium from Rats and Its Abrogation by Obesity is Mediated by the Leptin to Adiponectin Ratio. PLoS One 2016; 11: e0145992.

[134]

Hall ME, Harmancey R, Stec DE. Lean heart: Role of leptin in cardiac hypertrophy and metabolism. World J Cardiol 2015; 7: 511−524.

[135]

Kim HS, Kang JH, Jeung EB, Yang MP. Serum Concentrations of Leptin and Adiponectin in Dogs with Myxomatous Mitral Valve Disease. J Vet Intern Med 2016; 30: 1589−1600.

[136]

Hribal ML, Fiorentino TV, Sesti G. Role of C reactive protein (CRP) in leptin resistance. Curr Pharm Des 2014; 20: 609−615.

[137]

Genoux A, Bastard JP. Effects of leptin and adiponectin on the cardiovascular system. Ann Biol Clin (Paris) 2011; 78: 253−260.

[138]

Muse ED, Feldman DI, Blaha MJ, et al. The association of resistin with cardiovascular disease in the Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2015; 239: 101−108.

[139]

Mughal A, O'Rourke ST. Vascular effects of apelin: Mechanisms and therapeutic potential. Pharmacol Ther 2018; 190: 139−147.

[140]

Sabry MM, Mahmoud MM, Shoukry HS, et al. Interactive effects of apelin, renin-angiotensin system and nitric oxide in treatment of obesity-induced type 2 diabetes mellitus in male albino rats. Arch Physiol Biochem 2019; 125: 244−254.

[141]

January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol 2014; 64: e1−e76.

[142]

Kosztin A, Széplaki G, Kovács A, et al. Impact of CT-apelin and NT-proBNP on identifying non-responders to cardiac resynchronization therapy. Biomarkers 2017; 22: 279−286.

[143]

Chen YT, Wong LL, Liew OW, Richards AM. Heart Failure with Reduced Ejection Fraction (HFrEF) and Preserved Ejection Fraction (HFpEF): The Diagnostic Value of Circulating MicroRNAs. Cells 2019; 8: 1651.

[144]

Polyakova EA, Zaraiskii MI, Mikhaylov EN, et al. Association of myocardial and serum miRNA expression patterns with the presence and extent of coronary artery disease: A cross-sectional study. Int J Card 2020; 322: 9−15.

[145]

Gomes CPC, Schroen B, Kuster GM, et al. Regulatory RNAs in Heart Failure. Circulation 2020; 141: 313−328.

[146]

Qiang L, Hong L, Ningfu W, et al. Expression of miR-126 and miR-508–5p in endothelial progenitor cells is associated with the prognosis of chronic heart failure patients. Int J Cardiol 2013; 168: 2082−2088.

[147]

Ouyang S, Chen W, Zeng G, et al. MicroRNA-183–3p up-regulated by vagus nerve stimulation mitigates chronic systolic heart failure via the reduction of BNIP3L-mediated autophagy. Gene 2020; 726: 144136.

[148]

Vogel B, Keller A, Frese KS, et al. Multivariate miRNA signatures as biomarkers for non-ischaemic systolic heart failure. Eur Heart J 2013; 34: 2812−2822.

[149]

Tijsen AJ, Creemers EE, Moerland PD, et al. MiR423–5p as a circulating biomarker for heart failure. Circ Res 2010; 106: 1035−1039.

[150]

Ellis KL, Cameron VA, Troughton RW, et al. Circulating microRNAs as candidate markers to distinguish heart failure in breathless patients. Eur J Heart Fail 2013; 15: 1138−1147.

[151]

Corsten MF, Dennert R, Jochems S, et al. Circulating MicroRNA-208b and MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ Cardiovasc Genet 2010; 3: 499−506.

[152]

Vegter EL, van der Meer P, de Windt LJ, et al. MicroRNAs in heart failure: from biomarker to target for therapy. Eur J Heart Fail 2016; 18: 457−468.

[153]

Ovchinnikova ES, Schmitter D, Vegter EL, et al. Signature of circulating microRNAs in patients with acute heart failure. Eur J Heart Fail 2015; 18: 414−423.

[154]

Roncarati R, Viviani Anselmi C, Losi MA, et al. Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol 2014; 63: 920−927.

[155]

Ibrahim NE, Januzzi JL Jr. Established and Emerging Roles of Biomarkers in Heart Failure. Circ Res 2018; 123: 614−629.

[156]

Newgard CB. Metabolomics and metabolic diseases: Where do we stand? Cell Metab 2017; 25: 43−56.

[157]

Cheng S, Shah SH, Corwin EJ, et al. Potential Impact and Study Considerations of Metabolomics in Cardiovascular Health and Disease: A Scientific Statement From the American Heart Association. Circ Cardiovasc Genet 2017; 10: e000032.

[158]

Zheng Y, Yu B, Alexander D, et al. Associations between metabolomic compounds and incident heart failure among African Americans: the ARIC Study. Am J Epidemiol 2013; 178: 534−542.

[159]

Andersson C, Liu C, Cheng S, et al. Metabolomic signatures of cardiac remodelling andheart failure risk in the community. ESC Heart Fail 2020; 7: 3707−3715.

[160]

Barrett M, Boyne J, Brandts J, et al. Artificial intelligence supported patient self-care in chronic heart failure: a paradigm shift from reactive to predictive, preventive and personalised care. EPMA J 2019; 10: 445−464.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Published: 28 January 2021
Issue date: January 2021

Copyright

© 2021 JGC All rights reserved

Acknowledgements

ACKNOWLEDGEMENTS

The study has been supported by the grant from the Ministry of Science and Higher Education of the Russian Federation (agreement 075-15-2020-800). The authors declare no conflict of interests.

Rights and permissions

Return