Low level of neutrophil gelatinase-associated lipocalin (NGAL) in patients with chronic heart failure and multivessel coronary atherosclerosis

Year & Volume - Issue: 
Authors: 
Alla A. Garganeeva, Elena A. Kuzheleva, Vera A. Fediunina, Olga V. Tukish, Yuri V. Mareev, Marat Fudim, Sergey V. Popov
Heading: 
Article type: 
CID: 
e0102
PDF File: 
Abstract: 
Rationale — Several biomarkers are used to determine the prognosis of patients with heart failure (HF), including neutrophil gelatinase-associated lipocalin (NGAL, Lipocalin-2). We investigated NGAL serum levels in patients with HF and coronary artery disease (CAD). Methods — Fifty-three patients with chronic HF and stable multivessel CAD were included in the study. Patients were grouped on the basis of the NGAL level: low NGAL group (NGAL<37 ng/mL, n=19) and normal NGAL group (NGAL ≥37 ng/mL, n=34). Results and Discussion — The main findings from our study of patients with HF and multivessel CAD were: 1) Plasma NGAL levels were below reference values in 35.8% of cases. 2) We detected a strong positive correlation between neutrophil count and NGAL level (0.573, р<0.001). 3) Neutropenia was present in 15.8% of patients in the low NGAL group and in none in the group with a normal NGAL level (p=0.041). 4) Postinfarction left ventricular aneurysms were more often diagnosed in patients of the first group (15.8%) and in none of patients in the second group (p=0.041). Conclusion — Low NGAL has been associated with neutropenia in patients with heart failure and multivessel atherosclerotic CAD. Left ventricular aneurysms were diagnosed in 15.8% (n=3) patients with low NGAL and in none with normal NGAL levels. These changes may be due to maladaptive remodeling of the heart after myocardial infarction, but further research is needed.
Cite as: 
Garganeeva AA, Kuzheleva EA, Fediunina VA, Tukish OV, Mareev YuV, Fudim M, Popov SV. Low level of neutrophil gelatinase-associated lipocalin (NGAL) in patients with chronic heart failure and multivessel coronary atherosclerosis. Russian Open Medical Journal 2022; 11: e0102.

Introduction

Chronic heart failure (HF) is a severe progressive clinical syndrome [1-3]. Several biomarkers are used to determine the prognosis of patients with heart failure [3-9]. Neutrophil gelatinase-associated lipocalin (NGAL, Lipocalin-2) is one of such biomarkers [10]. It is synthesized in hepatocytes, cardiomyocytes, neurons, smooth muscle, renal, endothelial, and various populations of immune cells (e.g., neutrophils) [11]. Early clinical and experimental studies discovered NGAL levels closely associated with acute and chronic kidney disease [12-14]. Also, there is evidence that NGAL may be involved in the development of atherosclerotic vascular disease [15]. For example, NGAL content increased in patients with vulnerable carotid atherosclerotic plaques [16]. Other studies demonstrated that high NGAL levels were associated with vascular aneurysm rupture [17-18]. Also, NGAL could play a potential role in the development of abdominal aortic aneurysms [19]. High NGAL levels were associated with the development of early complications and mortality in patients with ST-segment elevation myocardial infarctions (STEMI) [20], along with acute and chronic heart failure [21-23]. In summary, most studies reported augmented levels of NGAL in patients with cardiovascular diseases, compared with healthy people. At the same time, some studies did not show NGAL as a predictor of clinical events in multivariate models following adjustment for renal function and brain natriuretic peptide (BNP) level [24-25]. However, there is a lack of clinical data on association low NGAL levels with cardiovascular pathology. Moreover, in a preclinical model, low NGAL was linked to adverse postinfarct remodeling of the left ventricle [26]. Therefore, we sought to investigate NGAL serum levels in patients with HF and coronary artery disease (CAD), scheduled for coronary artery bypass grafting (CABG).

 

Material and Methods

Study Design and Population

Fifty-three patients with chronic heart failure and stable multivessel CAD who were hospitalized for elective CABG were sequentially included in the study. The eligibility criteria were as follows: 1. Patients with chronic heart failure. 2. Triple vessel CAD revealed by invasive coronary angiography: atherosclerotic plaques ≥ 50% of the lumen of the vessel in the anterior descending, circumflex and right coronary arteries, or the presence of stenosis of the left coronary artery in combination with damage to the right coronary artery.

The exclusion criteria were as follows: severe respiratory failure (exacerbation of chronic obstructive pulmonary disease, uncontrolled course of bronchial asthma), diagnosed cancer, acute infection, exacerbation of chronic infectious disease, myocardial infarction, or progressive angina less than three months ago.

All patients were examined in accordance with a unified algorithm before surgery: review of anamnesis, clinical examination with determining body mass index (ratio of body weight in kilograms to the square of the body height in meters), symptoms and signs of heart failure. The instrumental examination included 12-lead electrocardiography (ECG), 24-hour Holter monitoring, and transthoracic echocardiography [27].

In addition, routine blood tests, including the absolute neutrophil count (reference values of neutrophils were 2-5.5×109/L, according to the local laboratory) were performed. The estimated glomerular filtration rate (eGFR) was calculated using the CKD-EPI equation. Coronary angiography was performed according to clinical indications within two months of inclusion in the study. The local institutional review board approved the study, and all patients provided written informed consent.

 

Biomarker Quantification

The concentration of serum NGAL was determined by enzyme-linked immunosorbent assay technique using Human NGAL Rapid ELISA Kit (KIT 037) (BioPorto Diagnostics, Denmark). Reference ranges of NGAL were 37-106 ng/mL, as per the manufacturer’s instructions (the limit based on the median value of plasma NGAL level in healthy individuals) [28].

 

Statistical Analyses

Data were analyzed with the use of STATISTICA 10.0 and R programming language in R studio software. Continuous variables were presented as median and interquartile range (IQR), considering the non-normal distribution parameters. Categorical data were presented in absolute and relative values: n (%). Continuous variables in the independent samples were analyzed using the Wilcoxon rank-sum test. The statistical significance of differences for categorical variables was determined using the two-sided Fisher’s exact test. The correlation between metrics was evaluated by calculating the Spearman correlation coefficient. A value of p<0.05 was considered statistically significant.

 

Results

The median serum concentration of NGAL was 41.6 (33.2; 51.7) ng/mL. There were no values greater than the upper reference value (106 ng/mL). Patients were grouped on the basis of the NGAL level: low NGAL group (NGAL<37 ng/mL, n=19) and normal NGAL group (NGAL≥37 ng/mL, n=34). Patients in the low NGAL group were older (on average, 67 vs. 59 years old, p=0.019) with no difference in sex ratio (Table 1). Previous myocardial infarctions were recorded in 58.8% of patients in the normal NGAL group and 73.7% in the low NGAL group (p=0.218).

 

Table 1. The clinical characteristics and medical history of patients with chronic heart failure and multivessel atherosclerosis of the coronary arteries vs. NGAL level

Parameter

Low NGAL group

(NGAL<37 ng/mL; n=19)

Normal NGAL group

(NGAL 37-106 ng/mL; n=34)

p-value

Men / women, n (%)

16/3 (84.2/15.8)

26/8 (76.5/23.5)

0.385

Age, years, Median (Q25; Q75),

67.0 (61.0; 68.0)

59.0 (55.0; 63.0)

0.019

Past myocardial infarction, n (%)

14 (73,7)

20 (58.8)

0.218

Time since last myocardial infarction Median (Q25; Q75), months

18.0 (5.0; 36.0)

12.0 (3.37; 78.0)

0.572

Hypertension, n (%)

19 (100)

32 (94.1)

0.407

Diabetes mellitus, n (%)

6 (31.6)

13 (38.2)

0.429

Arrhythmias, n (%)

12 (63.1)

13 (38.2)

0.072

Angina pectoris, n (%)

17 (89.5)

34 (100.0)

0.124

Dyspnea, n (%)

19 (100)

32 (94.1)

0.407

Palpitations, n (%)

12 (63.2)

7 (20.6)

0.003

Lower extremity edema, n (%)

2 (10.5)

6 (17.6)

0.395

Fatigue, n (%)

8 (42.1)

10 (29.4)

0.262

NGAL, neutrophil gelatinase-associated lipocalin; (Q25; Q75), interquartile range; p, significance level.

 

The severity of heart failure symptoms and exercise tolerance (sensu NYHA classification) were comparable between the groups (Table 2). There was no significant baseline difference in the main classes of cardiovascular medications (Table 3).

 

Table 2. Characterization of patients with heart failure

Parameter

Low NGAL group

(NGAL<37 ng/mL; n=19)

Normal NGAL group

(NGAL 37-106 ng/mL; n=34)

p-value

NT-proBNP, pg/ml, Median (Q25; Q75)

428(208;621)

397(219;580)

0.261

I NYHA, n (%)

0

3 (8.8)

0.255

II NYHA, n (%)

10 (52.6)

22 (64.7)

0.284

III NYHA, n (%)

9 (47.4)

9 (26.5)

0.108

IV NYHA, n (%)

0

0

 

HFpEF, n (%)

11 (57.9)

21 (67.7)

0.504

HFmrEF, n (%)

1 (5.3)

3 (8.8)

0.547

HFrEF, n (%)

7 (36.8)

8 (23.5)

0.236

NGAL, neutrophil gelatinase-associated lipocalin; NYHA, New York Heart Association functional classification; HFpEF, heart failure with preserved ejection fraction (≥50%); HFmrEF, heart failure with midrange ejection fraction (40-49%); HFrEF, heart failure with reduced ejection fraction (<40%); (Q25; Q75), interquartile range; p, significance level.

 

Table 3. Medical therapy in patients with chronic heart failure and multivessel atherosclerosis of the coronary arteries

Parameter

Low NGAL group

(NGAL<37 ng/mL; n=19)

Normal NGAL group

(NGAL 37-106 ng/mL; n=34)

p-value

Angiotensin-converting enzyme inhibitors, n (%)

7 (36.9)

15 (44.1)

0.413

Angiotensin II type receptor antagonists, n (%)

8 (42.1)

12 (35.3)

0.420

Beta-blockers, n (%)

15 (78.9)

33 (97)

0.106

Mineralocorticoid receptor antagonist, n (%)

6 (31.6)

2 (5.9)

0.019

Acetylsalicylic acid, n (%)

12 (63.2)

24 (70.6)

0.398

Clopidogrel, n (%)

6 (31.6)

12 (35.3)

0.510

Statins, n (%)

16 (84.2)

25 (73.5)

0.297

NGAL, neutrophil gelatinase-associated lipocalin; p, significance level.

 

Postinfarction left ventricular aneurysms were more often diagnosed in patients of the first group (15.8%) and in none of patients in the other group (p=0.041) (Table 4).

 

Table 4. Echocardiographic parameters in patients with chronic heart failure and multivessel atherosclerosis of the coronary arteries, depending on the level of NGAL

Parameter

Low NGAL group

(NGAL<37 ng/mL; n=19)

Normal NGAL group

(NGAL 37-106 ng/mL; n=34)

p-value

Left ventricular ejection fraction, %; Median (Q25; Q75)

50.0 (30.0;66.0)

61.5 (39.0;64.0)

0.385

Left ventricular aneurysm, n (%)

3 (15.8)

0 (0.0)

0.041

Left atrial volume index, mL/m2; Median (Q25; Q75)

46.8 (31.20;59.50)

35.25 (30.75;47.60)

0.151

Left ventricular end-diastolic volume, mL; Median (Q25; Q75)

130.5 (104.0;191.0)

117.5 (108.25;147.0)

0.939

Left ventricular end-systolic volume, mL; Median (Q25; Q75)

44.0 (37.0; 123.0)

51.0 (38.5; 84.5)

0.722

Stroke volume, mL Median (Q25; Q75)

67.0 (56.0;72.0)

69.5 (65.0; 74.5)

0.233

Left ventricular mass index, g/m2; Median (Q25; Q75)

97.0 (91.0;123.0)

95.5 (88.25;101.75)

0.470

E/A ratio; Median (Q25; Q75)

0.88 (0.57; 1.81)

0.87 (0.75; 1.23)

0.956

Е/e' ratio; Median (Q25; Q75)

8.18 (6.67;14.25)

9.25 (7.11; 11.18)

0.736

Moderate and severe mitral regurgitation, n (%)

1 (5.3)

3 (8.8)

0.547

Moderate and severe tricuspid regurgitation, n (%)

0

2 (5.9)

0.407

NGAL, neutrophil gelatinase-associated lipocalin; (Q25; Q75), interquartile range; p, significance level.

 

According to the results of a blood test, nearly every fifth patient with a low NGAL level (15.8%) was diagnosed with neutropenia, whereas in the group with normal NGAL values, the neutrophil level in all patients was within the reference range (p=0.041). Neutrophil level was missing in one patient. The absolute neutrophil count significantly differed between the groups as well: 2.74 (2.26; 3.46) and 4.03 (3.13; 5.17); p=0.001. There was a positive correlation between absolute neutrophil count and NGAL (r=0.573, р<0.001) (Figure 1).

 

Figure 1. Correlation between absolute neutrophil count and level of NGAL.

 

However, we did not find differences in the eGFR between the groups: 69.0 (61.0; 71.5) mL/min/1.73 m2 and 70.0 (60.0; 74.0) mL/min/1.73 m2 (p=0.602) (Table 5).

 

Table 5. Laboratory parameters in patients with chronic heart failure and multivessel atherosclerosis of the coronary arteries, depending on the level of NGAL

Parameter Median (Q25; Q75)

Group 1

(NGAL<37 ng/mL; n=19)

Group 2

(NGAL 37-106 ng/mL; n=34)

p-value

Hemoglobin, g/L

150.0 (137.0;162.0)

153.0 (142.0;166.0)

0.160

Red blood cell count, 1012/L

4.58 (4.22; 5.33)

4.94 (4.65;5.42)

0.083

Leukocytes, 109/L

6.5 (5.30;7.75)

8.15 (6.82; 9.32)

0.004

Neutrophil cell count, 109/L

2.74 (2.26;3.46)

4.03 (3.13;5.17)

0.001

Neutrophils, %

41.0 (36.22; 52.40)

49.7 (36.4; 51.7)

0.097

Monocytes, %

9.9 (8.95;11.32)

9.25 (8.0; 10.07)

0.107

Platelets, 109/L

202.5 (159.0; 225.25)

213.0 (190.75;258.0)

0.147

Total bilirubin, umol/L

17.25 (14.07;19.32)

17.55 (13.5;23.45)

0.860

Alanine aminotransferase, u/L

20.55 (15.17;27.90)

22.75 (16.40;26.45)

0.430

Aspartate aminotransferase, u/L

21.1 (18.00;24.60)

20.0 (15.7; 26.3)

0.682

Creatinine, mg/L

10.86 (9.67; 12.5)

10.86 (9.4; 11.99)

0.495

Blood urea, mg/dL

6.0 (4.9; 6.9)

5.9 (4.9; 6.5)

0.575

Glomerular filtration rate (CKD-EPI), ml/min/1.73m2

69.0 (61.0; 71.5)

70.0 (60.0; 74.0)

0.602

Sodium, mmol/L

142.3 (140.15; 143.85)

142.1 (140.1; 144.00)

0.719

Potassium, mmol/L

4.56 (4.45;5.15)

4.62 (4.21;4.88)

0.712

Protein, g/L

70.5 (68.05; 76.3)

72.4 (68.55;76.75)

0.904

Total cholesterol, mg/dL

3.86 (3.21; 4.55)

4.23 (3.72; 5.93)

0.297

Triglycerides, mg/dL

1.22 (1.04; 1.68)

1,78 (1.19;2.60)

0.002

Low-density lipoprotein, mg/dL

2.14 (0.91; 3.55)

2.72 (1.6; 4.03)

0.112

High-density lipoprotein, mg/dL

1.16 (1.03; 1.48)

1.15 (1.08; 1.24)

0.903

C-reactive protein, mg/L

4.3 (3.95;7.95)

4.5 (4.05;8.05)

0.423

NGAL, neutrophil gelatinase-associated lipocalin; (Q25; Q75), interquartile range; p, significance level.

 

Discussion

Our data represented the pioneering study of NGAL and cardiac structure/function in patients with heart failure and multivessel coronary atherosclerosis, who were scheduled for CABG. Previous studies in a similar cohort focused predominantly on renal outcomes in patients with different levels of NGAL after CABG [29-32].

The main findings from our study of patients with HF and multivessel CAD undergoing CABG were: 1) Plasma NGAL levels did not exceed reference values; moreover, NGAL levels were below reference values in 35.8% cases. 2) Since the NGAL level could be associated with the neutrophil count [33], we tested this association in our study and found a strong positive correlation between neutrophil count and NGAL level (0.573, р<0.001). 3) Neutropenia was present in 15.8 % of patients in the group with a low NGAL and none in the group with a normal NGAL level (p=0.041). 4) Postinfarction left ventricular aneurysms were more often diagnosed in patients of the first group (15.8%), and in none of patients in the other group (p=0.041), which could result from maladaptive remodeling of the heart after myocardial infarction [26]. It should be noted that the proportion of patients who had a history of myocardial infarction did not differ between the groups.

Earlier studies of NGAL demonstrated that low NGAL was more closely associated with neutropenia than with proinflammatory cytokines, procalcitonin and nutritional status, especially in patients with low levels of systemic inflammation while maintaining renal function [33].

Neutrophils matter in the healing process after myocardial infarction and subsequent remodeling of the heart. Yet, the exact role of neutrophils is ambiguous since both overactivation and insufficient activation may contribute to pathological remodeling of the heart and heart failure [34-38]. Mouse models demonstrated that neutrophil depletion during myocardial infarction led to excessive fibrosis, an increase in the left ventricular end-systolic volume, a significant decrease in left ventricular ejection fraction and cardiac output, which ultimately resulted in progressive deterioration of cardiac function [26]. One of the processes, required for adequate repair and cardiac remodeling, is macrophage efferocytosis, and its effectiveness depends on the level of NGAL [26, 39].

Accordingly, our results are consistent with experimental data on the association of low level of NGAL with features of post-infarction cardiac remodeling and development of heart failure. To date, there are no other convincing clinical studies demonstrating the association of low levels of NGAL with the clinical characteristics and medical history of patients with multivessel coronary atherosclerosis.

The main limitation of our study was its small sample size. Also, we had only cross-sectional data; and lack of follow-up information impaired our ability to draw conclusions about the interrelationship of NGAL and cardiac remodeling. Also, we did not know NGAL level at the time of acute myocardial infarction, leading to precipitating heart failure. Finally, clinical outcomes were not collected.

 

Conclusion

Low NGAL has been associated with neutropenia in patients with heart failure and multivessel atherosclerotic CAD. Left ventricular aneurysms were diagnosed in 15.8% (n=3) patients with low NGAL and in none with normal NGAL levels. These changes could be due to maladaptive remodeling of the heart after myocardial infarction, but further examination is needed. Further histological correlation between myocardial properties and NGAL, as well as study of the biomarker dynamics of NGAL after CABG, are needed.

 

Compliance with Ethical Standards

All performed procedures were in accordance with the ethical standards of the responsible committee on human experimentation (institutional and national) and with 1975 Declaration of Helsinki, year 2000 revision. Signed informed consent was obtained from all patients included in the study. The study was approved by the local institutional review board.

No animal studies were carried out by the authors for this article.

 

Funding

This study was supported by the state task, FSR № 122020300045-5.

 

Conflict of Interest

The authors declare that they have no conflicts of interest.

References: 
  1. Van der Meer P, Gaggin HK, Dec, GW. ACC/AHA versus ESC Guidelines on Heart Failure: JACC Guideline Comparison. J Am Coll Cardiol 2019; 73(21): 2756-2768. https://doi.org/10.1016/j.jacc.2019.03.478.
  2. Packer M. What causes sudden death in patients with chronic heart failure and a reduced ejection fraction? Eur Heart J 2019; 41(18): 1757-1763. https://doi.org/10.1093/eurheartj/ehz553.
  3. Mareev VY, Fomin IV, Ageev FT, Begrambekova YuL, Vasyuk YuA, Garganeeva AA, et al. Russian Heart Failure Society, Russian Society of Cardiology. Russian Scientific Medical Society of Internal Medicine Guidelines for Heart Failure: Chronic (CHF) and acute decompensated (ADHF). Diagnosis, prevention and treatment. Kardiologiia 2018; 58 Suppl 6: 8-158. Russian. https://doi.org/10.18087/cardio.2475.
  4. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, et. al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology. Eur Heart J 2016; 37(27): 2129-2200. https://doi.org/10.1093/eurheartj/ehw128.
  5. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DEJ, Colvin MM, et al. ACCF/ACC/HFSA focused update on new pharmacological therapy for heart failure: An update of the 2013 ACCF/AHA guidelines for the management of heart failure: A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines and the Heart Failure Society of America. J Am Coll Cardiol 2017; 70(6): 776-803. https://doi.org/10.1016/j.jacc.2017.04.025.
  6. Adamo L, Rocha-Resende C, Prabhu SD, Mann DL. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol 2020; 17(5): 269-285. https://doi.org/10.1038/s41569-019-0315-x.
  7. Aimo A, Castiglione V, Borrelli C, Saccaro LF, Franzini M, Stefano Masi S, et. al. Oxidative stress and inflammation in the evolution of heart failure: From pathophysiology to therapeutic strategies. Eur J Prev Cardiol 2019; 27(5): 494-510. https://doi.org/10.1177/2047487319870344.
  8. Carrillo-Salinas FJ, Ngwenyama N, Anastasiou M, Kaur K, Alcaide P. Heart inflammation: Immune cell roles and roads to the heart. Am J Pathol 2019; 189(8): 1482-1494. https://doi.org/10.1016/j.ajpath.2019.04.009.
  9. Mullens W, Damman K, Harjola VP, Mebazaa A, Brunner-La Rocca HP, Martens P, et. al. The use of diuretics in heart failure with congestion – A position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2019; 21(2): 137-155. https://doi.org/10.1002/ejhf.1369.
  10. Kjeldsen L, Johnsen AH, Sengeløv H, Borregaard N. Isolation and primary structure of NGAL, a novel protein associated with human neutrophil gelatinase. J Biol Chem 1993; 268(14): 10425-10432. https://pubmed.ncbi.nlm.nih.gov/7683678.
  11. Buonafine M, Martinez-Martinez E, Jaisser F. More than a simple biomarker: The role of NGAL in cardiovascular and renal diseases. Clin Sci (Lond) 2018; 132(9): 909-923. https://doi.org/10.1042/cs20171592.
  12. Khawaja S, Jafri L, Siddiqui I, Hashmi M, Ghani F. The utility of neutrophil gelatinase associated Lipocalin (NGAL) as a marker of acute kidney injury (AKI) in critically ill patients. Biomark Res 2019; 7: 4. https://doi.org/10.1186/s40364-019-0155-1.
  13. Zhang A, Cai Y, Wang PF, Qu JN, Luo ZC, Chen XD, et al. Diagnosis and prognosis of neutrophil gelatinase-associated lipocalin for acute kidney injury with sepsis: A systematic review and meta-analysis. Crit Care 2016; 20: 41. https://doi.org/10.1186/s13054-016-1212-x.
  14. Guo L, Zhu B, Yuan H, Zhao W. Evaluation of serum neutrophil gelatinase associated lipocalin in older patients with chronic kidney disease. Aging Med (Milton) 2020; 3(1): 32-39. https://doi.org/10.1002/agm2.12098.
  15. Shibata K, Sato K, Shirai R, Seki T, Okano T, Yamashita T, et al. Lipocalin-2 exerts pro-atherosclerotic effects as evidenced by in vitro and in vivo experiments. Heart Vessels 2020. https://doi.org/10.1007/s00380-020-01556-6.
  16. Eilenberg W, Stojkovic S, Kaider A, Piechota-Polanczyk A, Nanobachvili J, Domenig CM, et.al. Neutrophil gelatinase associated lipocalin (NGAL) for identification of unstable plaques in patients with asymptomatic carotid stenosis. Eur J Vasc Endovasc Surg 2019; 57(6): 768-777. https://doi.org/10.1016/j.ejvs.2018.12.029.
  17. Cione E, Piegari E, Gallelli G, Caroleo MC, Lamirata E, Curcio F, et al. Expression of MMP-2, MMP-9, and NGAL in tissue and serum of patients with vascular aneurysms and their modulation by statin treatment: A pilot study. Biomolecules 2020; 10(3): 359. https://doi.org/10.3390/biom10030359.
  18. Serra R, Grande R, Montemurro R, Butrico L, Caliò FG, Mastrangelo D, et al. The role of matrix metalloproteinases and neutrophil gelatinase-associated lipocalin in central and peripheral arterial aneurysms. Surgery 2015; 157(1): 155-162. https://doi.org/10.1016/j.surg.2014.06.008.
  19. Groeneveld ME, Struik JA, Musters RJP, Tangelder GJ, Koolwijk P, Niessen HW, et.al. The potential role of neutrophil gelatinase-associated lipocalin in the development of abdominal aortic aneurysms. Ann Vasc Surg 2019; 57: 210-219. https://doi.org/10.1016/j.avsg.2018.11.006.
  20. Al-Muhana SJ, Al-Mudhafa ZA, Bahlool R, Musa SI, Hadi NR. Impact of plasma neutrophil gelatinase-associated lipocalin level on early outcome of a patient with ST-segment elevation myocardial infarction. Int J Res Pharm 2019; 10(3), 1688-1691. https://doi.org/10.26452/ijrps.v10i3.1341.
  21. Maisel AS, Mueller C, Fitzgerald R, Brikhan R, Hiestand BC, Iqbal N, et al. Prognostic utility of plasma neutrophil gelatinase-associated lipocalin in patients with acute heart failure: The NGAL EvaLuation Along with B-type NaTriuretic Peptide in acutely decompensated heart failure (GALLANT) trial. Eur J Heart Fail 2011; 13(8): 846‐851. https://doi.org/10.1093/eurjhf/hfr087.
  22. Lábr K, Špinar J, Pařenica J, Špinarová L, Málek F, Špinarová M, et.al. Renal functions and prognosis stratification in chronic heart failure patients and the importance of neutrophil gelatinase-associated lipocalin. Kidney Blood Press Res 2018; 43(6): 1865-1877. https://doi.org/10.1159/000495819.
  23. Ito M, Doi K, Takahashi M, Koyama K, Myojo M. Hosoya Y, et.al. Plasma neutrophil gelatinase-associated lipocalin predicts major adverse cardiovascular events after cardiac care unit discharge. J Cardiol 2016; 67(2): 184-191. https://doi.org/10.1016/j.jjcc.2015.05.010.
  24. Shrestha K, Borowski AG, Troughton RW, Thomas JD, Klein AL, Tang WH. Renal dysfunction is a stronger determinant of systemic neutrophil gelatinase-associated lipocalin levels than myocardial dysfunction in systolic heart failure. J Card Fail 2011; 17(6): 472-478. https://doi.org/10.1016/j.cardfail.2011.02.003.
  25. Choi JW, Lee MH, Fujii T. Relationship between neutrophil gelatinase-associated lipocalin, cardiac biomarkers, inflammation index and renal parameters in cardiovascular disease. Arch Biol Sci 2019; 71(4): 621-628. https://doi.org/10.2298/ABS190604044C.
  26. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J 2017; 38(3): 187-197. https://doi.org/10.1093/eurheartj/ehw002.
  27. Mitchell C, Rahko PS, Blauwet LA, Canaday B, Finstuen JA, Foster MC, et al. Guidelines for performing a comprehensive transthoracic echocardiographic examination in adults: Recommendations from the American Society of Echocardiography. J Am Soc Echocardiogr 2019; 32(1): 1-64. https://doi.org/10.1016/j.echo.2018.06.004.
  28. Human NGAL Rapid ELISA Kit (KIT 037) For Human Urine, Plasma or Serum. Denmark: BioPorto Diagnostics. http://www.affinitydiagnostics.ca/pdf/_250_NGAL%20Rapid%20ELISA%20Brochure.pdf.
  29. Miah OF, Dowel FA, Latif A, Hai AN, Mahmud MA, Razzak MA, et al. NGAL (neutrophil gelatinase-associated lipocalin) is an early predictor of acute kidney injury after cardiac surgery and variation of NGAL values in homogenous study subject. Mymensingh Med J 2018; 27(1): 212-215. https://pubmed.ncbi.nlm.nih.gov/29459617.
  30. Saeedi Ghaheh M, Mardani S, Malekpour A, Kadkhodaei Elyaderani F, Salehi Choliche F, Mortazavi N, et al. Comparison of urinary KIM-1 and NGAL and plasma creatinine in patients undergoing coronary artery bypass graft surgery. J Nephropharmacol 2018; 10(1): eo4. https://doi.org/10.34172/npj.2021.04.
  31. Kanchi M, Manjunath R, Massen J, Vincent L, Belani K. Neutrophil gelatinase-associated lipocalin as a biomarker for predicting acute kidney injury during off-pump coronary artery bypass grafting. Ann Card Anaesth 2017; 20(3): 297-302. https://doi.org/10.4103/aca.ACA_48_17.
  32. Balkanay OO, Göksedef D, Ömeroğlu SN, İpek G. The Reliability of the Use of serum neutrophil gelatinase-associated lipocalin levels in the assessment of renal functions after coronary artery bypass grafting. Cardiol Res Pract 2018; 2018: 7291254. https://doi.org/10.1155/2018/7291254.
  33. Choi JW, Fujii T. The Prevalence of low plasma neutrophil gelatinase-associated lipocalin level in systemic inflammation and its relationship with proinflammatory cytokines, procalcitonin, nutritional status, and leukocyte profiles. Clin Lab 2019; 65(6). https://doi.org/10.7754/clin.lab.2019.181235.
  34. Kain V, Halade GV. Role of neutrophils in ischemic heart failure. Pharmacol Ther 2020; 205: 107424. https://doi.org/10.1016/j.pharmthera.2019.107424.
  35. Peiseler M, Kubes P. More friend than foe: The emerging role of neutrophils in tissue repair. J Clin Invest 2019; 129(7): 2629-2639. https://doi.org/10.1172/JCI124616.
  36. Strassheim D, Dempsey EC, Gerasimovskaya E, Stenmark K, Karoor V. Role of inflammatory cell subtypes in heart failure. J Immunol Res 2019; 2019: 2164017. https://doi.org/10.1155/2019/2164017.
  37. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: From inflammation to fibrosis. Circ Res 2016; 119(1): 91-112. https://doi.org/10.1161/circresaha.116.303577.
  38. Tourki B, Halade G. Leukocyte diversity in resolving and nonresolving mechanisms of cardiac remodeling. FASEB J 2017; 31(10): 4226-4239. https://doi.org/10.1096/fj.201700109r.
  39. Lawrence SM, Corriden R, Nizet V. How neutrophils meet their end. Trends Immunol 2020; 41(6): 531-544. https://doi.org/10.1016/j.it.2020.03.008.
About the Authors: 

Alla A. Garganeeva – MD, DSc, Professor, Head of the Department of Myocardial Pathology, Cardiology Research Institute, Tomsk National Research Medical Center, Tomsk, Russia. https://orcid.org/0000-0002-9488-6900
Elena A. Kuzheleva – MD, PhD, Researcher, Department of Myocardial Pathology, Cardiology Research Institute, Tomsk National Research Medical Center, Tomsk, Russia. https://orcid.org/0000-0002-8070-2234
Vera A. Fediunina – Junior Researcher, Department of Myocardial Pathology, Cardiology Research Institute, Tomsk National Research Medical Center, Tomsk, Russia. https://orcid.org/0000-0003-1437-8250
Olga V. Tukish – MD, PhD, Researcher, Department of Myocardial Pathology, Cardiology Research Institute, Tomsk National Research Medical Center, Russia. https://orcid.org/0000-0002-7661-5808
Yuri V. Mareev – MD, PhD, Researcher, National Medical Research Center for Therapy and Preventive Medicine, Moscow, Russia; Robertson Center For Biostatistics, Glasgow, United Kingdom. https://orcid.org/0000-0002-1939-7189
Marat Fudim – Medicinae Doctor, Master of Health Science Degree, Duke University Medical Center, Division of Cardiology, Durham, NC, USA. https://orcid.org/0000-0002-8671-7007
Sergey V. Popov – MD, DSc, Professor, Academician of the Russian Academy of Sciences, Director of the Research Institute of Cardiology, Tomsk National Research Medical Center, Tomsk, Russia. https://orcid.org/0000-0002-9050-4493

Received 17 November 2021, Accepted 27 January 2022 
© 2021, Russian Open Medical Journal 
Correspondence to Olga V. Tukish. Department of Myocardial Pathology, Cardiology Research Institute, Tomsk National Research Medical center, Russian Academy of Sciences, Russia, Kievskaya, 111-A, Tomsk, 634012 Russia. Phone: + 73822565275. E-mail: olgatukish@yandex.ru.

DOI: 
10.15275/rusomj.2022.0102