Next Article in Journal
Sexual Behaviour and Fantasies in a Group of Young Italian Cohort
Next Article in Special Issue
Walking the Line with Ticagrelor: Meta-Analysis Comparing the Safety and Efficacy of Ticagrelor Monotherapy after a Short Course of Ticagrelor-Based Dual Antiplatelet Therapy versus Standard Therapy in Complex Percutaneous Coronary Intervention
Previous Article in Journal
Mortality, Risk Factors and Risk Assessment after Periprosthetic Femoral Fractures—A Retrospective Cohort Study
Previous Article in Special Issue
Clinical Value and Time Course of Pericoronary Fat Inflammation in Patients with Angiographically Nonobstructive Coronaries: A Preliminary Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Brief Report

Association between Serum Osteoprotegerin Levels and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction

1
Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France
2
Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(19), 4326; https://doi.org/10.3390/jcm10194326
Submission received: 16 August 2021 / Revised: 13 September 2021 / Accepted: 17 September 2021 / Published: 23 September 2021
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Coronary Artery Disease)

Abstract

:
Background. Osteoprotegerin (OPG), a glycoprotein of the tumour necrosis factor (TNF) superfamily, is one of the main biomarkers for vascular calcification. Aim. We aimed to evaluate the association between serum OPG levels and extent of coronary lesions in patients with acute myocardial infarction (MI). Methods. Consecutive patients hospitalized for an acute MI who underwent coronary angiography were included. SYNTAX score was calculated to assess the severity of coronary artery disease. The population was analysed in low (5 (3–6)), medium (11 (9–13)) and high (20 (18–23)) tertiles of SYNTAX score. Results. Among the 378 patients included, there was a gradual increase in age, rate of diabetes, anterior wall location, and a reduction in left ventricular ejection fraction across the SYNTAX tertiles. OPG levels significantly increased across the tertiles (962 (782–1497), 1240 (870–1707), and 1464 (1011–2129) pg/mL, respectively (p < 0.001)). In multivariate analysis, OPG [OR(CI95%): 2.10 (1.29–3.49) 0.003], were associated with the high SYNTAX group, beyond hypercholesterolemia, CV history and reduced glomerular filtration rate. Conclusion. We found an association between OPG levels and coronary lesions complexity patients with acute MI.

1. Introduction

Osteoprotegerin (OPG), a glycoprotein of the tumour necrosis factor (TNF) receptor superfamily, acts as a dummy receptor for the nuclear factor κ-B ligand receptor (RANKL) and the TNF-linked apoptosis-inducing ligand (TRAIL) [1,2]. It is expressed in vivo in bone cells (osteoblasts), vascular smooth muscle cells and endothelial cells [3,4]. It is detected by immunohistochemistry in aortic and coronary atherosclerotic plaques. OPG is known to inhibit osteoclastogenesis and animal studies have shown a protective effect [5]. Vascular calcification (VC) is an active, complex process, where the key step towards calcification is the transformation of vascular smooth muscle cells to osteoblast phenotype, subsequently leading to hydroxyapatite accumulation, and the formation of cartilage and bone in the arterial wall. Although the underlying mechanisms linking VC to atherosclerotic cardiovascular diseases (ASCVD) are largely unknown, growing evidence suggests that OPG plays a key role in the development of VC [6]. High levels of OPG have been associated with coronary artery calcification and the development of coronary artery disease (CAD) [7,8,9,10,11]. Moreover, serum OPG could be considered a useful biomarker in patients with acute or chronic cardiometabolic disease [12].
However, only a few studies have evaluated the relationship between the extent of CAD by coronary angiography and circulating levels of OPG. Jono et al. showed an association between serum OPG and the number of coronary lesions in a series of 200 patients with stable CAD [10]. However, the clinical significance of OPG levels in patients with acute MI remains unexplored.
The objective of our study was to address the relationship between serum OPG levels and the extension of CAD in patients with acute MI, as measured by the severity of the coronary lesions using the SYNTAX score.

2. Materials and Methods

2.1. Population

From the RICO survey (RICO) [13], all consecutive patients admitted in the coronary care unit of Dijon University Hospital for an acute MI from May 2017 to February 2018 and who underwent coronary angiography were prospectively included. Patients < 18 y, with prior AF (paroxysmal or permanent), type 2 MI or time delay from symptom onset to admission > 12H were excluded from the study. MI was defined according to current criteria of European Society of Cardiology (ESC) [14,15]. All of the participants provided consent prior to inclusion, and the Ethics Committee of the University Hospital of Dijon approved the protocol (BIOCARDIS-2016–9205AAO034S02117).

2.2. Data Collection

CV risk factors were collected, i.e., age, gender, hypertension, diabetes, obesity (body mass index (BMI)), prior high cholesterol or total cholesterol > 2.5 g/L, familial history of CAD (premature CAD in a first-degree relative < 55 years of age for men or <65 years of age for women) and current smoking.
ASCVD history (CAD, stroke, carotid atheroma or peripheral arterial disease (PAD)), chronic renal failure or congestive heart failure were also collected. Systolic and diastolic blood pressure (SBP and DBP), heart rate (HR) were measured on admission. Biological data were obtained from blood sampling on admission (blood glucose, CRP, NT-pro-BNP, creatinine and estimated Glomerular Filtration Rate (CKD-EPI). Troponin Ic peak was obtained from 3 blood samples every 8 h after admission. Left ventricular ejection fraction (LVEF) was measured within 24 h using the Simpson biplane method [16]. GRACE risk score was also calculated [17]. In-hospital events including CV mortality, heart failure (as defined as Killip class >2) and recurrent MI were collected.

2.3. Serum OPG Level Assessment

Blood samples were taken upon admission to assess circulating OPG levels from radial or femoral artery in a non-heparinized test tube. Blood samples were stored at +4 °C for up to 24 h. Stability of the biomarker in preclinical conditions has been tested in healthy volunteers (n = 10) and in MI patients (n = 6). The samples were centrifuged and stored at −80 °C before analysis. For each assay, a pooled sample from 3 healthy volunteers was used as a control. Serum OPG was measured in duplicate using a commercially available multiplex assay (Human Magnetic Luminex Assay, R&D Systems—Bio-Techne, Lille, France), according to manufacturer instructions. The detection limit of this assay was (25 pg/mL) and both the intra-and interassay coefficients of variation were < 10%.

2.4. Coronary Angiography

Coronary angiography images were reviewed blinded (without knowledge of the patient’s group) by two trained interventional cardiologists. A discrepancy was noted for 7 cases, for which a joint review allowed the final adjudication. SYNTAX anatomical score, before PCI was calculated to address CAD extent and coronary lesions’ complexity (length, bifurcation, diffuse disease, calcification, thrombus, total occlusion) [17].

2.5. Statistical Analysis

Dichotomous variables were expressed as n (%) and continuous variables in median (interquartile range (IQR)). The normality of the variables was tested by the Kolmogorov–Smirnoff test. For categorical data, a chi-square or Fischer’s exact test and a Student’s test or a Mann–Whitney test for continuous data were used, as appropriate. The significance threshold was set at 5%. A multivariate logistic regression analysis was performed to estimate high SYNTAX score (last tertile), including variables significantly associated in univariate analysis, with an inclusion and exclusion threshold set at 5% (Age > 65 year, hypertension, diabetes, hypercholesterolemia, prior CAD, GFR < 60 mL/min/1.73 m2). The optimal OPG threshold (>1080 pg/mL) for predicting a high SYNTAX score was obtained by the ROC (receiver–operation characteristic) curve analysis (AUC 0.60, p < 0.001) and Youden’s index (sensitivity: 58%; specificity: 70%).

3. Results

Of the 791 consecutive patients admitted for a heart attack during the inclusion period, 378 were analysed (Figure 1).
Baseline characteristics are presented on SYNTAX score tertiles, ranging from low (5(3–6)), to medium (11(9–13)) and high (20(18–23)) SYNTAX score (Table 1). Across the tertiles, there was a gradual increase in age, rate of diabetes, anterior wall location and GRACE risk score, while LVEF was gradually reduced. The rate of STEMI/NSTEMI was similar across the tertiles (p = 0.520). Patients from the highest SYNTAX tertile had more prior ASCVD and CABG than in lower tertiles. In addition, hospital CV events were more frequent in the highest SYNTAX group (Table 1).
Patients with more severe CAD extent (Tertile 3) had higher levels of OPG (p < 0.001) (Figure 2) and rates of patients with high OPG (>1080 pg/mL) gradually increased across the tertiles (Table 2).
Patients with a high SYNTAX score had also elevated creatinine (p < 0.001), and NT-pro-BNP levels (p < 0.001). However, the Troponin Ic peak was similar for the three groups.
In multivariate analysis, a high OPG level was the strongest factor associated with high CAD extent (OR: 2.09(1.31–3.32)), beyond hypercholesterolemia, prior CAD and reduced GFR (Table 3).

4. Discussion

Our work on a large prospective study in acute MI highlights that circulating levels of OPG are associated with the severity of CAD, as assessed by the SYNTAX score.
Vascular calcifications (VC) are typical features of advanced ASCVD, and OPG, as a bone key for atherosclerosis, is associated with disease progression. Moreover, patients with VC are at higher risk for CV events [18]. A growing body of evidence suggests that the triad OPG and its ligands (RANKL and TRAIL) exert pro-atherogenic and pro-diabetogenic effects by amplifying the adverse effects of inflammation [19]. Moreover, an increase in serum OPG levels had been shown to be predictor of CV mortality in patients with chronic coronary syndrome [20,21]. A 10-year follow-up survey showed that serum OPG levels were an independent risk factor for the progression of atherosclerosis, CV events and mortality [22]. Finally, OPG has been shown to predict early carotid atherosclerosis in patients with CAD [23,24].

4.1. OPG Is Associated with the Severity of Coronary Artery Disease

Serum OPG levels increased with the number of lesions on coronary angiography in 200 patients with CAD [11]. More recently, serum OPG levels correlated with CAD severity by angiographic Gensini score, and CV mortality was higher at midterm follow-up in patients with high OPG levels [25]. In our study, we further showed a strong association between the degree of complexity of the coronary lesions by SYNTAX score and the OPG level in patients with acute MI. In contrast, in a prior study including 40 patients, no significant association had been found between serum OPG levels and elevated SYNTAX scores [13]. However, the small sample size and the high number of exclusion criteria weaken the conclusions drawn from this work.
Our work showed a strong and independent relationship between OPG levels and the severity of CAD in acute MI. Therefore, this biomarker, which is easily measured with blood sampling on admission, could help in predicting the severity of CAD.

4.2. OPG as a New Biomarker in Coronary Artery Disease

We showed that patients with elevated serum OPG (≥1080 pg/mL) were more likely to have a high SYNTAX score. An upper limit of OPG at 1412 pg/mL with an AUC of 0.60 (p = 0.028) was found to be related with the calcium score in patients with chronic kidney disease [25]. Moreover, a significant association between the OPG level and a calcium score >100, found an OPG cut-off value of 953 pg/mL with a sensitivity of 62% and a specificity of 82% [26]. These thresholds, close to those of our current work, could be proposed as a reference limit beyond which we could consider ischemia testing in patients at high CV risk. In agreement with the literature, we found that CV risk factors, such as hypercholesterolemia, CV history and elevated creatinine level, were strong predictors of extensive CAD [27,28,29]. Given the potential therapeutic strategies targeting VC, further studies are thus needed to better understand the place of OPG as a biomarker in ASCVD [30].

4.3. Study Limitations

This study has several limitations. This was a monocentric study and our findings may not be extrapolated to other MI populations. Moreover, coronary calcium score, a reliable surrogate marker of atherosclerosis, was not available in our study. However, characteristics including the median age (63 years) and the rate of main CV risk factors are similar to those found in MI registries, strongly suggesting their representativeness [31]. Finally, pre-analytical variables may influence OPG measurements. However, to reduce variability within the process, all the pre-analytical and analytical conditions were similar for all study participants, as recommended [32].

5. Conclusions

In a prospective study on patients with acute MI, we found an association between serum OPG levels and CAD burden, as assessed by the SYNTAX score. Further multicentre studies are needed to confirm the relevance of the determination of OPG levels in patients with acute MI.

Author Contributions

Conceptualization, Y.C. and R.I.; methodology, M.B. and B.M.; formal analysis, B.M.; investigation, C.V. and L.R.; resources, A.M., C.V, F.B. and L.R.; data curation, L.T.; writing—original draft preparation, M.Z.; writing—review and editing, supervision, M.Z.; project administration, Y.C. and C.V.; funding acquisition, Y.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the University Hospital of Dijon, the Association de Cardiologie de Bourgogne, Fédération Française de Cardiologie, and by grants from the Agence Régionale de Santé de Bourgogne Franche-Comté, and from the Conseil Régional de Bourgogne Franche-Comté.

Institutional Review Board Statement

Ethics Committee of the University Hospital of Dijon approved the protocol (BIOCARDIS-2016–9205AAO034S02117).

Data Availability Statement

Supporting data may be found in internal archives.

Acknowledgments

The authors thank Suzanne Rankin for English review, Sylvie Mazencieux-Agobert for editing assistance, Mailis Saint Jalmes, Jessica Massenot and Etienne Baudouin for their help in data collection.

Conflicts of Interest

Y.C.: consultant or speaker for Bayer, BMS/Pfizer, Boehringer Ingelheim, Novartis, Sanofi and Servier. M.Z. received research grants from Amarin Corp.

References

  1. Collin-Osdoby, P. Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ. Res. 2004, 95, 1046–1057. [Google Scholar] [CrossRef]
  2. Hofbauer, L.C.; Khosla, S.; Dunstan, C.R.; Lacey, D.L.; Boyle, W.J.; Riggs, B.L. The roles of osteoprotegerin and osteoprotegerin ligand in the paracrine regulation of bone resorption. J. Bone Miner. Res. 2000, 15, 2–12. [Google Scholar] [CrossRef]
  3. Emery, J.G.; McDonnell, P.; Burke, M.B.; Deen, K.C.; Lyn, S.; Silverman, C.; Dul, E.; Appelbaum, E.R.; Eichman, C.; DiPrinzio, D.; et al. Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. J. Biol. Chem. 1998, 273, 14363–14367. [Google Scholar] [CrossRef] [Green Version]
  4. Schoppet, M.; Preissner, K.T.; Hofbauer, L.C. RANK ligand and osteoprotegerin: Paracrine regulators of bone metabolism and vascular function. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 549–553. [Google Scholar] [CrossRef] [Green Version]
  5. Hao, Y.; Tsuruda, T.; Sekita-Hatakeyama, Y.; Kurogi, S.; Kubo, K.; Sakamoto, S.; Nakamura, M.; Udagawa, N.; Sekimoto, T.; Hatakeyama, K.; et al. Cardiac hypertrophy is exacerbated in aged mice lacking the osteoprotegerin gene. Cardiovasc. Res. 2016, 110, 62–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Harper, E.; Forde, H.; Davenport, C.; Rochfort, K.D.; Smith, D.; Cummins, P.M. Vascular calcification in type-2 diabetes and cardiovascular disease: Integrative roles for OPG, RANKL and TRAIL. Vascul. Pharmacol. 2016, 82, 30–40. [Google Scholar] [CrossRef] [PubMed]
  7. Kiechl, S.; Schett, G.; Wenning, G.; Redlich, K.; Oberhollenzer, M.; Mayr, A.; Santer, P.; Smolen, J.; Poewe, W.; Willeit, J. Osteoprotegerin is a risk factor for progressive atherosclerosis and cardiovascular disease. Circulation 2004, 109, 2175–2180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Vik, A.; Mathiesen, E.B.; Brox, J.; Wilsgaard, T.; Njølstad, I.; Jørgensen, L.; Hansen, J.B. Serum osteoprotegerin is a predictor for incident cardiovascular disease and mortality in a general population: The Tromsø Study. J. Thromb. Haemost. 2011, 9, 638–644. [Google Scholar] [CrossRef]
  9. Abedin, M.; Omland, T.; Ueland, T.; Khera, A.; Aukrust, P.; Murphy, S.A.; Jain, T.; Gruntmanis, U.; McGuire, D.K.; de Lemos, J.A. Relation of osteoprotegerin to coronary calcium and aortic plaque (from the Dallas Heart Study). Am. J. Cardiol. 2007, 99, 513–518. [Google Scholar] [CrossRef]
  10. Jono, S.; Ikari, Y.; Shioi, A.; Mori, K.; Miki, T.; Hara, K.; Nishizawa, Y. Serum osteoprotegerin levels are associated with the presence and severity of coronary artery disease. Circulation 2002, 106, 1192–1194. [Google Scholar] [CrossRef] [Green Version]
  11. Schoppet, M.; Sattler, A.M.; Schaefer, J.R.; Herzum, M.; Maisch, B.; Hofbauer, L.C. Increased osteoprotegerin serum levels in men with coronary artery disease. J. Clin. Endocrinol. Metab. 2003, 88, 1024–1028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Rochette, L.; Meloux, A.; Rigal, E.; Zeller, M.; Cottin, Y.; Vergely, C. The role of osteoprotegerin in the crosstalk between vessels and bone: Its potential utility as a marker of cardiometabolic diseases. Pharmacol. Ther. 2018, 182, 115–132. [Google Scholar] [CrossRef] [PubMed]
  13. Putot, A.; Chague, F.; Manckoundia, P.; Cottin, Y.; Zeller, M. Post-infectious Myocardial Infarction: New Insights for Improved Screening. J. Clin. Med. 2019, 8, 827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ibanez, B.; James, S.; Agewall, S.; Antunes, M.J.; Bucciarelli-Ducci, C.; Bueno, H.; Caforio, A.L.P.; Crea, F.; Goudevenos, J.A.; Halvorsen, S.; et al. ESC Scientific Document GroupESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation. Eur. Heart J. 2018, 39, 119–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Collet, J.P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur. Heart J. 2020, 29, ehaa575. [Google Scholar] [CrossRef]
  16. Donal, E.; Place, C.D. Étude de la fonction systolique ventriculaire gauche par échocardiographie Doppler: Anciennes et nouvelles approches. MT Cardio. 2006, 2, 329–338. [Google Scholar]
  17. Kappetein, A.P.; Dawkins, K.D.; Mohr, F.W.; Morice, M.C.; Mack, M.J.; Russell, M.E.; Pomar, J.; Serruys, P.W.J.C. Current percutaneous coronary intervention and coronary artery bypass grafting practices for three-vessel and left main coronary artery disease. Insights from the SYNTAX run-in phase. Eur. Assoc. Cardiothorac. Surg. 2006, 29, 486–491. [Google Scholar] [CrossRef] [Green Version]
  18. Nakahara, T.; Dweck, M.R.; Narula, N.; Pisapia, D.; Narula, J.; Strauss, H.W. Coronary Artery Calcification: From Mechanism to Molecular Imaging. JACC Cardiovasc. Imaging 2017, 10, 582–593. [Google Scholar] [CrossRef]
  19. Lee, G.L.; Yeh, C.C.; Wu, J.Y.; Lin, H.C.; Wang, Y.F.; Kuo, Y.Y.; Hsieh, Y.T.; Hsu, Y.J.; Kuo, C.C. TLR2 Promotes Vascular Smooth Muscle Cell Chondrogenic Differentiation and Consequent Calcification via the Concerted Actions of Osteoprotegerin Suppression and IL-6-Mediated RANKL Induction. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 432–445. [Google Scholar] [CrossRef] [PubMed]
  20. Jono, S.; Otsuki, S.; Higashikuni, Y.; Shioi, A.; Mori, K.; Hara, K.; Hashimoto, H.; Ikari, Y. Serum osteoprotegerin levels and long-term prognosis in subjects with stable coronary artery disease. J. Thromb. Haemost. 2010, 8, 1170–1175. [Google Scholar] [CrossRef]
  21. Venuraju, S.M.; Yerramasu, A.; Corder, R.; Lahiri, A. Osteoprotegerin as a Predictor of Coronary Artery Disease and Cardiovascular Mortality and Morbidity. J. Am. Coll. Cardiol. 2010, 55, 2049–2061. [Google Scholar] [CrossRef] [Green Version]
  22. Kiechl, S.; Werner, P.; Knoflach, M.; Furtner, M.; Willeit, J.; Schett, G. The osteoprotegerin/RANK/RANKL system: A bone key to vascular disease. Expert. Rev. Cardiovasc. Ther. 2006, 4, 801–811. [Google Scholar] [CrossRef]
  23. Montecucco, F.; Steffens, S.; Mach, F. The immune response is involved in atherosclerotic plaque calcification: Could the RANKL/RANK/OPG system be a marker of plaque instability? Clin. Dev. Immunol. 2007, 2007, 75805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Morisawa, T.; Nakagomi, A.; Kohashi, K.; Kosugi, M.; Kusama, Y.; Atarashi, H.; Shimizu, W. Osteoprotegerin is Associated With Endothelial Function and Predicts Early Carotid Atherosclerosis in Patients With Coronary Artery Disease. Int. Heart J. 2015, 56, 605–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ghaffari, S.; Yaghoubi, A.; Baghernejad, R.; Sepehrvand, N.; Sokhanvar, S.; Haghjou, A.G. The value of serum osteoprotegerin levels in patients with angina like chest pain undergoing diagnostic coronary angiography. Cardiol. J. 2013, 20, 261–267. [Google Scholar] [CrossRef] [Green Version]
  26. Morena, M.; Dupuy, A.-M.; Jaussent, I.; Vernhet, H.; Gahide, G.; Klouche, K.; Bargnoux, A.-S.; Delcourt, C.; Canaud, B.; Cristol, J.-P. A cut-off value of plasma osteoprotegerin level may predict the presence of coronary artery calcifications in chronic kidney disease patients. Nephrol. Dial. Transplant. 2009, 24, 3389–3397. [Google Scholar] [CrossRef] [Green Version]
  27. Homorodean, C.; Iancu, A.C.; Dregoesc, I.M.; Spînu, M.; Ober, M.C.; Tãtaru, D.; Leucuta, D.; Olinic, M. Renal Failure Impact on the Outcomes of ST-Segment Elevation Myocardial Infarction Patients Due to a Left Main Coronary Culprit Lesion Treated Using a Primary Percutaneous Coronary Intervention. J. Clin. Med. 2019, 8, 565. [Google Scholar] [CrossRef] [Green Version]
  28. Lin, T.; Wang, L.; Guo, J.; Liu, P.; Chen, L.; Wei, M.; Li, G. Association Between Serum LDL-C and ApoB and SYNTAX Score in Patients With Stable Coronary Artery Disease. Angiology 2018, 69, 724–729. [Google Scholar] [CrossRef] [PubMed]
  29. Xu, J.-J.; Song, Y.; Jiang, P.; Gao, Z.; Li, J.-X.; Chen, J.; Yang, Y.-J.; Gao, R.-L.; Xu, B.; Yuan, J.-Q. Effect of prior stroke on long-term outcomes of percutaneous coronary interventions in Chinese patients: A large single-center study. Catheter. Cardiovasc. Interv. 2019, 93, E75–E80. [Google Scholar] [CrossRef]
  30. Rochette, L.; Meloux, A.; Rigal, E.; Zeller, M.; Malka, G.; Cottin, Y.; Vergely, C. The Role of Osteoprotegerin in Vascular Calcification and Bone Metabolism: The Basis for Developing New Therapeutics. Calcif. Tissue Int. 2019, 105, 239–251. [Google Scholar] [CrossRef]
  31. Puymirat, E.; Cayla, G.; Cottin, Y.; Elbaz, M.; Henry, P.; Gerbaud, E.; Lemesle, G.; Popovic, B.; Labèque, J.N.; Roubille, F.; et al. Twenty-year trends in profile, management and outcomes of patients with ST-segment elevation myocardial infarction according to use of reperfusion therapy: Data from the FAST-MI program 1995–2015. Am. Heart J. 2019, 214, 97–106. [Google Scholar] [CrossRef] [PubMed]
  32. De Ciriza, C.P.; Lawrie, A.; Varo, N. Influence of pre-analytical and analytical factors on osteoprotegerin measurements. Clin. Biochem. 2014, 47, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow chart.
Figure 1. Flow chart.
Jcm 10 04326 g001
Figure 2. Median OPG levels according to SYNTAX score (p < 0.001). Circle: outlier; *: extreme oulier.
Figure 2. Median OPG levels according to SYNTAX score (p < 0.001). Circle: outlier; *: extreme oulier.
Jcm 10 04326 g002
Table 1. Baseline characteristics, according to SYNTAX score tertiles. n(%) or median (IQR).
Table 1. Baseline characteristics, according to SYNTAX score tertiles. n(%) or median (IQR).
SYNTAX Score
Tertile 1Tertile 2Tertile 3p
N = 125N = 128N = 125
SYNTAX score5 (3–6)11 (9–13)20 (18–23)<0.001
Min_max2–78–1617–36
Risk factors
Age (year)58 (53–71)64 (56–74)69 (58–83)<0.001
  Age > 65 year53 (42%)67 (52%)83 (66%)<0.001
BMI (kg/m2)26 (24–29)27 (24–30)26 (24–30)0.404
Women44 (35%)39 (31%)35 (28%)0.514
Hypertension63 (50%)74 (58%)81 (65%)0.051
Diabetes26 (21%)29 (23%)44 (35%)0.016
Hypercholesterolemia48 (38%)48 (38%)67 (53%)0.013
Family history of CAD31 (24%)31 (24%)22 (18%)0.333
Current smoking55 (43%)32 (25%)31 (25%)0.001
CV history
Chronic coronary syndrome36% (28%)54 (42%)43 (34%)0.067
Prior MI16 (13%)12 (9%)20 (16%)0.284
CABG0 (0%)0 (0%)6 (5%)0.002
PCI18 (14%)13 (10%)16 (13%)0.612
ASCVD32 (25%)20 (16%)46 (37%)0.001
Chronic renal failure7 (6%)5 (4%)9 (7%)0.518
Congestive heart failure2 (2%)0 (0%)1 (1%)0.364
Admission data
Time to admission (min)148 (93–440)153 (89–383)170 (80–329)0.729
HR (b/min)77 (63–88)78 (65–90)78 (70–91)0.024
SBP (mmHg)143 (128–168)147 (126–170)134 (114–165)0.209
SBP (mmHg)89 (77–96)87 (74–100)79 (65–100)0.113
LVEF (%)60 (50–60)58 (50–60)50 (45–55)<0.001
  LVEF < 40%5 (4%)10 (8%)16 (13%)0.029
STEMI64 (50%)73 (57%)70 (56%)0.520
Anterior wall location11 (9%)23 (18%)40 (32%)<0.001
GRACE risk score128 (105–155)140 (112–161)150 (133–178)0.028
Revascularization
PCI88 (70%)123 (96%)88 (70%)<0.001
CABG0 (0%)1 (1%)29 (23%)<0.001
In-hospiral events
CV death2 (2%)1 (1%)9 (7%)0.006
Heart failure5 (4%)7 (6%)15 (12%)0.030
Recurrent MI1 (1%)1 (1%)3 (2%)0.430
ASCVD: atherosclerotic cardiovascular disease; PCI: percutaneous coronary intervention; CABG: coronary artery bypass graft; CAD: coronary artery disease; BMI: body mass index; STEMI: ST segment elevation myocardial infarction; LVEF: left ventricular ejection fraction; SBP: systolic blood pressure; DBP: diastolic blood pressure; HR: heart rate.
Table 2. Biological data. N(%) or median(IQR).
Table 2. Biological data. N(%) or median(IQR).
SYNTAX Score
Tertile 1Tertile 2Tertile 3p-Value
N = 125N = 128N = 125
OPG, pg/mL962 (782–1497)1240 (870–1707)1464 (1011–2129)<0.001
  OPG >1080 pg/mL46 (23%)70 (35%)82 (41%)<0.001
Creatinine (μmol/L)75 (61–92)77 (68–91)82 (69–97)0.002
  GFR(mL/min/1.73 m2)93 (77–101)85 (71–97)78 (59–95)<0.001
Total cholesterol (mmol/L)5.11 (4.59–6.08)5.42 (4.51–6.21)5.06 (3.89–5.99)0.094
HDL cholesterol (mmol/L)1.23 (1.04–1.58)1.32 (1.03–1.55)1.27 (1.00–1.50)0.577
LDL cholesterol (mmol/L)3.20 (2.38–3.82)3.29 (2.50–4.11)3.05 (2.07–3.88)0.151
Triglycerides (mmol/L)1.34 (0.89–2.10)1.53 (1.02–2.16)1.40 (0.98–2.00)0.518
Blood glucose (mmol/L)7.1 (5.9–8.8)6.9 (5.8–8.4)7.1 (6.2–9.0)0.016
CRP (mg/L)4.3 (2.9–8.9)3.0 (2.9–9.9)4.1 (2.9–10.2)0.027
Ntpro-BNP (pg/mL)135 (50–1015)183 (80–1120)511 (104–1829)<0.001
Troponine Ic peak (μg/L)30.5 (8.2–78.8)22.0 (4.5–93.8)34.5 (4.7–157.5)0.246
OPG: osteoprotegerin; Nt-proBNP: N-terminal proBrain Natriuretic peptide; CRP: C-reactive protein; LDL: low density lipoprotein; HDL: high density lipoprotein.
Table 3. Multivariate logistic regression analysis for the prediction of CAD extent (last SYNTAX Tertile) (OR: odds ratio; CI: confidence interval).
Table 3. Multivariate logistic regression analysis for the prediction of CAD extent (last SYNTAX Tertile) (OR: odds ratio; CI: confidence interval).
OR (95% CI)p
OPG > 1080 pg/mL2.10 (1.29–3.49)0.003
Hypercholesterolemia1.69 (1.08–2.89)0.019
Prior CAD1.47 (1.11–4.56)0.018
GFR < 60 mL/min/1.73 m21.99 (1.20–3.05)0.002
GFR: glomerular filtration rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cottin, Y.; Issa, R.; Benalia, M.; Mouhat, B.; Meloux, A.; Tribouillard, L.; Bichat, F.; Rochette, L.; Vergely, C.; Zeller, M. Association between Serum Osteoprotegerin Levels and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction. J. Clin. Med. 2021, 10, 4326. https://doi.org/10.3390/jcm10194326

AMA Style

Cottin Y, Issa R, Benalia M, Mouhat B, Meloux A, Tribouillard L, Bichat F, Rochette L, Vergely C, Zeller M. Association between Serum Osteoprotegerin Levels and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction. Journal of Clinical Medicine. 2021; 10(19):4326. https://doi.org/10.3390/jcm10194326

Chicago/Turabian Style

Cottin, Yves, Rany Issa, Mourad Benalia, Basile Mouhat, Alexandre Meloux, Laura Tribouillard, Florence Bichat, Luc Rochette, Catherine Vergely, and Marianne Zeller. 2021. "Association between Serum Osteoprotegerin Levels and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction" Journal of Clinical Medicine 10, no. 19: 4326. https://doi.org/10.3390/jcm10194326

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop