1
|
Mishra PP, Mishra BH, Lyytikäinen LP, Goebeler S, Martiskainen M, Hakamaa E, Kleber ME, Delgado GE, März W, Kähönen M, Karhunen PJ, Lehtimäki T. Genetic risk score for coronary artery calcification and its predictive ability for coronary artery disease. Am J Prev Cardiol 2024; 20:100884. [PMID: 39483246 PMCID: PMC11525111 DOI: 10.1016/j.ajpc.2024.100884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/08/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
Aim The modest added predictive value of the existing genetic risk scores (GRSs) for coronary artery disease (CAD) could be partly due to missing genetic components, hidden in the genetic architecture of intermediate phenotypes such as coronary artery calcification (CAC). In this study, we investigated the predictive ability of CAC GRS for CAD. Materials and methods We investigated the association of CAC GRSs with CAD and coronary calcification among the participants in the Ludwigshafen Risk and Cardiovascular Health study (LURIC) (n = 2742), the Tampere Vascular Study (TVS) (n = 133), and the Tampere Sudden Death Study (TSDS) (n = 660) using summary data from the largest multi-ancestry GWAS meta-analysis of CAC to date. Added predictive value of the CAC GRS over the traditional CVD risk factors as well as metaGRS, a GRS for CAD constructed with 1.7 million genetic variants, was tested with standard train-test machine learning approach using the LURIC data, which had the largest sample size. Results CAC GRS was significantly associated with CAD in LURIC (OR=1.41, 95 % CI [1.28-1.55]), TVS (OR=1.79, 95 % CI [1.05-3.21]) as well as in TSDS (OR=4.20, 95 % CI [1.74-10.52]). CAC GRS showed strong association with calcification areas in left (OR=1.78, 95 % CI [1.16-2.74]) and right (OR=1.71, 95 % CI [1.98-2.67]) coronary arteries. There was statistically significant added predictive value of the CAC GRS for CAD over the used traditional CVD risk factors (AUC 0.734 vs 0.717, p-value = 0.02). Furthermore, CAC GRS improved the prediction accuracy for CAD when combined with metaGRS. Conclusions This study showed that CAC GRS is a new risk marker for CAD in three European cohorts, with added predictive value over the traditional CVD risk factors.
Collapse
Affiliation(s)
- Pashupati P. Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Binisha H. Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Sirkka Goebeler
- Forensic Medicine, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mika Martiskainen
- Forensic Medicine, Finnish Institute for Health and Welfare, Helsinki, Finland
- Faculty of Medicine and Health Technology, Tampere University and Fimlab Laboratories, Tampere, Finland
| | - Emma Hakamaa
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcus E. Kleber
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Graciela E. Delgado
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere Finland
| | - Pekka J. Karhunen
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
2
|
Enroth M, Sievanen H, Asikainen TTMA, Viik J. Use of ST/HR hysteresis decreases false positive rate in exercise electrocardiography test of middle-aged asymptomatic women. J Electrocardiol 2024; 87:153820. [PMID: 39504597 DOI: 10.1016/j.jelectrocard.2024.153820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Exercise electrocardiography (ECG) is a common diagnostic and prognostic method for the detection of coronary artery disease (CAD). However, its accuracy in asymptomatic women has not been comprehensively investigated and the diagnostic criteria may require refinements. This study evaluated the performance of ECG-derived ST/HR-index, ST/HR hysteresis and ST-segment depression parameters among asymptomatic middle-aged women. METHODS 108 women (mean age 56 ± 4 years) performed exercise ECG test on treadmill until exhaustion three times within a nine-month period. False-positive rates of maximum ST/HR-index, ST/HR hysteresis, and ST-segment depression values measured from 12 leads at peak exercise and after one-minute recovery were evaluated with recommended diagnostic partition values. Repeatability was assessed with intraclass correlation (ICC) and Bland-Altman plot analysis. RESULTS False-positive rate was lower for all variables when a two‑lead configuration was used instead of a single‑lead configuration. Using a two‑lead configuration, ST/HR hysteresis (0-1.9 %) and ST-segment depression after one-minute recovery (0-2.8 %) had lower false-positive rates compared to ST/HR index (3.7-20.4 %) and ST-segment depression at peak exercise (9.3-27.8 %). ICC values indicated moderate repeatability for ST/HR hysteresis while ST/HR index, ST-segment at peak exercise, and ST-segment after one-minute recovery had moderate-to-good repeatability. Bland-Altman analysis indicated poor repeatability for all evaluated ECG variables. CONCLUSION In asymptomatic middle-aged women, practitioners should prefer the use of ST/HR hysteresis and ST-segment after one-minute recovery over the conventional ST-segment depression at peak exercise or ST/HR index, and evaluate the ECG data from a two‑lead configuration instead of single‑lead.
Collapse
Affiliation(s)
- Miro Enroth
- Faculty of Medicine, Riga Stradins University, Riga, Latvia.
| | - Harri Sievanen
- The UKK Institute for Health Promotion Research, Tampere, Finland
| | | | - Jari Viik
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
3
|
Hernesniemi JA, Pukkila T, Molkkari M, Nikus K, Lyytikäinen LP, Lehtimäki T, Viik J, Kähönen M, Räsänen E. Prediction of Sudden Cardiac Death With Ultra-Short-Term Heart Rate Fluctuations. JACC Clin Electrophysiol 2024; 10:2010-2020. [PMID: 38878016 DOI: 10.1016/j.jacep.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Conventional measures of heart rate variability (HRV) have shown only modest associations with sudden cardiac death (SCD). Detrended fluctuation analysis (DFA), with novel methodological developments to evaluate the short-term scaling exponent, is a potentially superior method compared to conventional HRV tools. OBJECTIVES In this study, the authors studied the analysis of the association between DFA and SCD. METHODS The investigators studied the predictive value of ultra-short-term heart rate fluctuations (1-minute electrocardiogram samples) with DFA at rest and during different stages of physical exertion for incident SCD among 2,794 participants undergoing clinical exercise testing in the prospective FINCAVAS (Finnish Cardiovascular Study). The novel key DFA measure, the short-scale scaling exponent computed with second-order detrending (DFA2 α1), was the main exposure variable. SCDs were defined by American Heart Association/European Society of Cardiology criteria using death certificates with written accounts of the events. RESULTS During a median follow-up of 8.3 years (Q1-Q3: 6.4-10.5), 83 SCDs occurred. DFA2 α1 measured at rest (but not in exercise) associated highly significantly with the risk of SCD, with 1-SD lower values associating with a 2.4-fold (Q1-Q3: 2.0-3.0) risk (P < 0.001). The results persisted when adjusting for other major risk factors for SCD, including age, cardiovascular morbidities, cardiorespiratory fitness, heart rate reduction, and left ventricular ejection fraction. Associations between conventional HRV parameters (measured at any stage of exercise or at rest) and SCD were substantially weaker and statistically nonsignificant after adjusting for other risk factors. CONCLUSIONS Ultra-short-term DFA2 α1, when measured at rest, is a powerful and independent predictor of SCD. The association between DFA2 α1 and SCD is modified by physical exertion.
Collapse
Affiliation(s)
- Jussi A Hernesniemi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland; Heart Hospital, Tampere University Hospital, Tampere, Finland.
| | - Teemu Pukkila
- Computational Physics Laboratory, Tampere University, Tampere, Finland
| | - Matti Molkkari
- Computational Physics Laboratory, Tampere University, Tampere, Finland
| | - Kjell Nikus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland; Heart Hospital, Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland; Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Jari Viik
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland
| | - Mika Kähönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Tampere, Finland; Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland
| | - Esa Räsänen
- Computational Physics Laboratory, Tampere University, Tampere, Finland
| |
Collapse
|
4
|
The role of β-adrenergic stimulation in QT interval adaptation to heart rate during stress test. PLoS One 2023; 18:e0280901. [PMID: 36701349 PMCID: PMC9879473 DOI: 10.1371/journal.pone.0280901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
The adaptation lag of the QT interval after heart rate (HR) has been proposed as an arrhythmic risk marker. Most studies have quantified the QT adaptation lag in response to abrupt, step-like changes in HR induced by atrial pacing, in response to tilt test or during ambulatory recordings. Recent studies have introduced novel methods to quantify the QT adaptation lag to gradual, ramp-like HR changes in stress tests by evaluating the differences between the measured QT series and an estimated, memoryless QT series obtained from the instantaneous HR. These studies have observed the QT adaptation lag to progressively reduce when approaching the stress peak, with the underlying mechanisms being still unclear. This study analyzes the contribution of β-adrenergic stimulation to QT interval rate adaptation in response to gradual, ramp-like HR changes. We first quantify the QT adaptation lag in Coronary Artery Disease (CAD) patients undergoing stress test. To uncover the involved mechanisms, we use biophysically detailed computational models coupling descriptions of human ventricular electrophysiology and β-adrenergic signaling, from which we simulate ventricular action potentials and ECG signals. We characterize the adaptation of the simulated QT interval in response to the HR time series measured from each of the analyzed CAD patients. We show that, when the simulated ventricular tissue is subjected to a time-varying β-adrenergic stimulation pattern, with higher stimulation levels close to the stress peak, the simulated QT interval presents adaptation lags during exercise that are more similar to those measured from the patients than when subjected to constant β-adrenergic stimulation. During stress test recovery, constant and time-varying β-adrenergic stimulation patterns render similar adaptation lags, which are generally shorter than during exercise, in agreement with results from the patients. In conclusion, our findings support the role of time-varying β-adrenergic stimulation in contributing to QT interval adaptation to gradually increasing HR changes as those seen during the exercise phase of a stress test.
Collapse
|
5
|
Beyene SD, Nikus KC, Lehtimäki TJ, Kähönen MAP, Viik JJ. Evaluation of the QRS score for diagnosing coronary artery disease in women: A Finnish cardiovascular study. Ann Noninvasive Electrocardiol 2022; 27:e12968. [PMID: 35580147 PMCID: PMC9296785 DOI: 10.1111/anec.12968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/12/2022] [Accepted: 04/25/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Exercise electrocardiography is a widely used diagnostic modality for diagnosing coronary artery disease. This method has been used for both sexes; however, its diagnostic accuracy in women is limited. METHODS The study analyzed 332 women participating in the Finnish Cardiovascular Study. Among 332 women, 125 with angiographically proven coronary artery disease (mean age 62.1 ± 9.5 years), 91 with a low likelihood of coronary artery disease (mean age 47.3 ± 13.5 years), and 116 without angiographically proven coronary artery disease (mean age 56.3 ± 9.9 years) were analyzed. The Q, R, S, and ST-segment changes and QRS score were determined by subtracting the Q, R, S, and ST-segment amplitudes immediately after the maximal exercise changes from their rest values (Δ). Receiver operating characteristic curve analysis was performed to evaluate the overall diagnostic performance of the parameters for predicting coronary artery disease. RESULTS The areas under the receiver operating characteristic curve between coronary artery disease and low likelihood of coronary artery disease groups for the QRS score and ΔSTV5, ΔQaVF, and ΔRaVF were 0.75, 0.73, 0.71, and 0.71, respectively. These areas were lower (0.62, 0.57, 0.60, and 0.60, respectively) between the groups with and without angiographically proven coronary artery disease. QRS score demonstrated the highest sensitivity at 80% specificity (61.5%) and the highest specificity at 80% sensitivity (57.6%). CONCLUSIONS This study suggests that the QRS and ST-segment depression have a moderate diagnostic ability to predict coronary artery disease in women. Q and R waves in lead aVF showed good diagnostic ability.
Collapse
Affiliation(s)
- Serkalem D. Beyene
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | | | - Terho J. Lehtimäki
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Mika A. P. Kähönen
- Department of Clinical PhysiologyTampere University Hospital, and Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Jari J. Viik
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| |
Collapse
|
6
|
Del Castillo MG, Hernando D, Orini M, Laguna P, Viik J, Bailón R, Pueyo E. QT variability unrelated to RR variability during stress testing for identification of coronary artery disease. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200261. [PMID: 34689618 DOI: 10.1098/rsta.2020.0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 06/13/2023]
Abstract
Stress test electrocardiogram (ECG) analysis is widely used for coronary artery disease (CAD) diagnosis despite its limited accuracy. Alterations in autonomic modulation of cardiac electrical activity have been reported in CAD patients during acute ischemia. We hypothesized that those alterations could be reflected in changes in ventricular repolarization dynamics during stress testing that could be measured through QT interval variability (QTV). However, QTV is largely dependent on RR interval variability (RRV), which might hinder intrinsic ventricular repolarization dynamics. In this study, we investigated whether different markers accounting for low-frequency (LF) oscillations of QTV unrelated to RRV during stress testing could be used to separate patients with and without CAD. Power spectral density of QTV unrelated to RRV was obtained based on time-frequency coherence estimation. Instantaneous LF power of QTV and QTV unrelated to RRV were obtained. LF power of QTV unrelated to RRV normalized by LF power of QTV was also studied. Stress test ECG of 100 patients were analysed. Patients referred to coronary angiography were classified into non-CAD or CAD group. LF oscillations in QTV did not show significant differences between CAD and non-CAD groups. However, LF oscillations in QTV unrelated to RRV were significantly higher in the CAD group as compared with the non-CAD group when measured during the first phases of exercise and last phases of recovery. ROC analysis of these indices revealed area under the curve values ranging from 61 to 73%. Binomial logistic regression analysis revealed LF power of QTV unrelated to RRV, both during the first phase of exercise and last phase of recovery, as independent predictors of CAD. In conclusion, this study highlights the importance of removing the influence of RRV when measuring QTV during stress testing for CAD identification and supports the added value of LF oscillations of QTV unrelated to RRV to diagnose CAD from the first minutes of exercise. This article is part of the theme issue 'Advanced computation in cardiovascular physiology: new challenges and opportunities'.
Collapse
Affiliation(s)
| | - David Hernando
- I3A, University of Zaragoza, IIS Aragón, Spain
- CIBER-BBN, Zaragoza, Spain
| | | | - Pablo Laguna
- I3A, University of Zaragoza, IIS Aragón, Spain
- CIBER-BBN, Zaragoza, Spain
| | - Jari Viik
- Tampere University of Technology, Tampere, Finland
| | - Raquel Bailón
- I3A, University of Zaragoza, IIS Aragón, Spain
- CIBER-BBN, Zaragoza, Spain
| | - Esther Pueyo
- I3A, University of Zaragoza, IIS Aragón, Spain
- CIBER-BBN, Zaragoza, Spain
| |
Collapse
|
7
|
Laaksonen J, Mishra PP, Seppälä I, Lyytikäinen LP, Raitoharju E, Mononen N, Lepistö M, Almusa H, Ellonen P, Hutri-Kähönen N, Juonala M, Raitakari O, Kähönen M, Salonen JT, Lehtimäki T. Examining the effect of mitochondrial DNA variants on blood pressure in two Finnish cohorts. Sci Rep 2021; 11:611. [PMID: 33436758 PMCID: PMC7804469 DOI: 10.1038/s41598-020-79931-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
High blood pressure (BP) is a major risk factor for many noncommunicable diseases. The effect of mitochondrial DNA single-nucleotide polymorphisms (mtSNPs) on BP is less known than that of nuclear SNPs. We investigated the mitochondrial genetic determinants of systolic, diastolic, and mean arterial BP. MtSNPs were determined from peripheral blood by sequencing or with genome-wide association study SNP arrays in two independent Finnish cohorts, the Young Finns Study and the Finnish Cardiovascular Study, respectively. In total, over 4200 individuals were included. The effects of individual common mtSNPs, with an additional focus on sex-specificity, and aggregates of rare mtSNPs grouped by mitochondrial genes were evaluated by meta-analysis of linear regression and a sequence kernel association test, respectively. We accounted for the predicted pathogenicity of the rare variants within protein-encoding and the tRNA regions. In the meta-analysis of 87 common mtSNPs, we did not observe significant associations with any of the BP traits. Sex-specific and rare-variant analyses did not pinpoint any significant associations either. Our results are in agreement with several previous studies suggesting that mtDNA variation does not have a significant role in the regulation of BP. Future studies might need to reconsider the mechanisms thought to link mtDNA with hypertension.
Collapse
Affiliation(s)
- Jaakko Laaksonen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland.
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Maija Lepistö
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Nina Hutri-Kähönen
- Department of Paediatrics, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland.,Division of Medicine, Turku University Hospital, Turku, Finland.,Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.,Research Centre for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka T Salonen
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,MAS-Metabolic Analytical Services Oy, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| |
Collapse
|
8
|
Orlandi M, Orlandi G, Bini V, Fiorillo C, Becatti M, Stefani L. The ST segment depression pattern in asymptomatic peri-menopausal female athletes. Heliyon 2020; 6:e04738. [PMID: 32923714 PMCID: PMC7475225 DOI: 10.1016/j.heliyon.2020.e04738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022] Open
Abstract
Background ST segment has not been well investigated in asymptomatic peri-menopausal female athletes, when the CV risk is higher. Aims The aims of the study is to investigate the prevalence of ST segment depression in peri-menopausal female athletes, divided in four age groups. Methods in a cohort of 6010 female athletes aged 45–65 years old, 161 subjects were selected for the presence of ST segment depression, revealed by maximal ergometric test. All athletes were also evaluated by physical examination and echocardiography. Inclusion criteria for ST segment depression were ST depression >0.5 mm respect to baseline and its depth was divided in 0.5 mm, 0.5–1 mm, 1–2 mm categories. Its behavior was classified in ascending, horizontal and descending and it was studied in relation to the age range (<51; 51–55; 56–60; >60). Results ST segment depression was mainly evident in inferolateral leads in all groups (63%, 70,3%, 71%, 63,6%, for <51; 51–55, 56–60 and 61–65 respectively) with horizontal pattern (52,2%, 59,5%, 57,8%, 63,6%) and 1–2 mm depth (43,5%, 46,7%, 60,6%), with the exception of the range 51–55, mainly showing a depth of 0,5-1 mm (45,9%). The older group showed increased SBP (137.5 mmHg, p = 0.007) and BMI (24.3, p = 0.093) values. Mitral prolapse was shown in 11,8% while 36,6% showed systolic flattening of mitral leaflets. Conclusions ST depression in asymptomatic menopausal female athletes is frequent and it is characterized by a specific presentation pattern. This is particularly important in the menopausal age when CV risk factors are more prevalent.
Collapse
Affiliation(s)
| | | | - Vittorio Bini
- Department of Medicine, University of Perugia, Italy
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Italy
| | - Laura Stefani
- Sports Medicine Center, University of Florence, Italy
| |
Collapse
|
9
|
Mahmoodi BK, Tragante V, Kleber ME, Holmes MV, Schmidt AF, McCubrey RO, Howe LJ, Direk K, Allayee H, Baranova EV, Braund PS, Delgado GE, Eriksson N, Gijsberts CM, Gong Y, Hartiala J, Heydarpour M, Pasterkamp G, Kotti S, Kuukasjärvi P, Lenzini PA, Levin D, Lyytikäinen LP, Muehlschlegel JD, Nelson CP, Nikus K, Pilbrow AP, Tang W, van der Laan SW, van Setten J, Vilmundarson RO, Deanfield J, Deloukas P, Dudbridge F, James S, Mordi IR, Teren A, Bergmeijer TO, Body SC, Bots M, Burkhardt R, Cooper-DeHoff RM, Cresci S, Danchin N, Doughty RN, Grobbee DE, Hagström E, Hazen SL, Held C, Hoefer IE, Hovingh GK, Johnson JA, Kaczor MP, Kähönen M, Klungel OH, Laurikka JO, Lehtimäki T, Maitland-van der Zee AH, McPherson R, Palmer CN, Kraaijeveld AO, Pepine CJ, Sanak M, Sattar N, Scholz M, Simon T, Spertus JA, Stewart AFR, Szczeklik W, Thiery J, Visseren FL, Waltenberger J, Richards AM, Lang CC, Cameron VA, Åkerblom A, Pare G, März W, Samani NJ, Hingorani AD, ten Berg JM, Wallentin L, Asselbergs FW, Patel R. Association of Factor V Leiden With Subsequent Atherothrombotic Events: A GENIUS-CHD Study of Individual Participant Data. Circulation 2020; 142:546-555. [PMID: 32654539 PMCID: PMC7493828 DOI: 10.1161/circulationaha.119.045526] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Studies examining the role of factor V Leiden among patients at higher risk of atherothrombotic events, such as those with established coronary heart disease (CHD), are lacking. Given that coagulation is involved in the thrombus formation stage on atherosclerotic plaque rupture, we hypothesized that factor V Leiden may be a stronger risk factor for atherothrombotic events in patients with established CHD. METHODS We performed an individual-level meta-analysis including 25 prospective studies (18 cohorts, 3 case-cohorts, 4 randomized trials) from the GENIUS-CHD (Genetics of Subsequent Coronary Heart Disease) consortium involving patients with established CHD at baseline. Participating studies genotyped factor V Leiden status and shared risk estimates for the outcomes of interest using a centrally developed statistical code with harmonized definitions across studies. Cox proportional hazards regression models were used to obtain age- and sex-adjusted estimates. The obtained estimates were pooled using fixed-effect meta-analysis. The primary outcome was composite of myocardial infarction and CHD death. Secondary outcomes included any stroke, ischemic stroke, coronary revascularization, cardiovascular mortality, and all-cause mortality. RESULTS The studies included 69 681 individuals of whom 3190 (4.6%) were either heterozygous or homozygous (n=47) carriers of factor V Leiden. Median follow-up per study ranged from 1.0 to 10.6 years. A total of 20 studies with 61 147 participants and 6849 events contributed to analyses of the primary outcome. Factor V Leiden was not associated with the combined outcome of myocardial infarction and CHD death (hazard ratio, 1.03 [95% CI, 0.92-1.16]; I2=28%; P-heterogeneity=0.12). Subgroup analysis according to baseline characteristics or strata of traditional cardiovascular risk factors did not show relevant differences. Similarly, risk estimates for the secondary outcomes including stroke, coronary revascularization, cardiovascular mortality, and all-cause mortality were also close to identity. CONCLUSIONS Factor V Leiden was not associated with increased risk of subsequent atherothrombotic events and mortality in high-risk participants with established and treated CHD. Routine assessment of factor V Leiden status is unlikely to improve atherothrombotic events risk stratification in this population.
Collapse
Affiliation(s)
- Bakhtawar K. Mahmoodi
- St. Antonius Hospital, department of Cardiology, Koekoekslaan 1, 3435CM, Nieuwegein, the Netherlands
- Division of Hemostasis and Thrombosis, Department of Hematology, UMC Groningen, University of Groningen, Groningen, the Netherlands
| | - Vinicius Tragante
- Department of Cardiology, Division Heart and Lungs, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Michael V. Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
| | - Amand F. Schmidt
- Department of Cardiology, Division Heart and Lungs, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
| | - Raymond O. McCubrey
- Intermountain Heart Institute, Intermountain Medical Center, Salt Lake City, UT, USA
| | - Laurence J. Howe
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
| | - Kenan Direk
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
| | - Hooman Allayee
- Departments of Preventive Medicine and Biochemistry and Molecular Medicine, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Ekaterina V. Baranova
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, the Netherlands
| | - Peter S. Braund
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | | | | | - Yan Gong
- University of Florida, Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, 1333 Center Drive, Gainesville, FL 32608, USA
| | - Jaana Hartiala
- Departments of Preventive Medicine and Biochemistry and Molecular Medicine, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
- Institute for Genetic Medicine, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Mahyar Heydarpour
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Gerard Pasterkamp
- Department of Clinical Chemistry, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Salma Kotti
- Assistance Publique-Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, Platform of Clinical Research of East Paris (URCEST-CRCEST-CRB HUEP-UPMC), Paris, France
| | - Pekka Kuukasjärvi
- Department of Cardio-Thoracic Surgery, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Techonology, Tampere University, Arvo Ylpön katu 34, Tampere 33014, Finland
| | - Petra A. Lenzini
- Washington University School of Medicine, Department of Genetics, Statistical Genomics Division, Saint Louis, Missouri, USA
| | - Daniel Levin
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Arvo Ylpön katu 34, Tampere 33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Techonology, Tampere University, Tampere 33014, Finland
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Ensitie 4, 33520 Tampere, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33014, Finland
| | - Anna P. Pilbrow
- The Christchurch Heart Institute, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
| | - W.H.Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institue, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jessica van Setten
- Department of Cardiology, Division Heart and Lungs, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ragnar O. Vilmundarson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - John Deanfield
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London Medical School, Queen Mary University of London, London, UK
- Centre for Genomic Health, Queen Mary University of London, London, UK
| | - Frank Dudbridge
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Stefan James
- Uppsala Clinical Research Center, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Ify R Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Andrej Teren
- Heart Center Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Thomas O. Bergmeijer
- St. Antonius Hospital, department of Cardiology, Koekoekslaan 1, 3435CM, Nieuwegein, the Netherlands
| | - Simon C. Body
- Department of Anaesthesiology, Boston University School of Medicine, 750 Albany St, Boston, MA 02118, USA
| | - Michiel Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Ralph Burkhardt
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Rhonda M. Cooper-DeHoff
- University of Florida, Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, 1333 Center Drive, Gainesville, FL 32608, USA
- College of Medicine, Division of Cardiovascular Medicine, University of Florida, 1600 SW Archer Road/Box 100277, Gainesville, FL 32610, USA
| | - Sharon Cresci
- Washington University School of Medicine, Department of Genetics, Statistical Genomics Division, Saint Louis, Missouri, USA
- Washington University School of Medicine, Department of Medicine, Cardiovascular Division, Saint Louis, Missouri, USA
| | - Nicolas Danchin
- Assistance Publique-Hôpitaux de Paris (APHP), Department of Cardiology, Hôpital Européen Georges Pompidou, 75015 Paris, France; FACT (french Alliance for cardiovascular trials); Université Paris Descartes, Paris, France
- Université Paris-Descartes, Paris, France
| | - Robert N. Doughty
- Heart Health Research Group, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Diederick E. Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala University, Dept of Cardiology, Uppsala, Sweden and Uppsala Clinical Research Center, Uppsala, Sweden
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institue, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, and Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Claes Held
- Uppsala Clinical Research Center, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Imo E. Hoefer
- Department of Clinical Chemistry and Hematology, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - G. Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Julie A. Johnson
- University of Florida, Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, 1333 Center Drive, Gainesville, FL 32608, USA
- College of Medicine, Division of Cardiovascular Medicine, University of Florida, 1600 SW Archer Road/Box 100277, Gainesville, FL 32610, USA
| | - Marcin P. Kaczor
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str, 31-066 Kraków, Poland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, FM1 3rd floor, Tampere 33521, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Techonology, Tampere University, Tampere 33014, Finland
| | - Olaf H. Klungel
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, the Netherlands
| | - Jari O. Laurikka
- Department of Cardio-Thoracic Surgery, Heart Center, Tampere University Hospital, Arvo Ylpön katu 6, Tampere 33521, Finland
- Department of Cardio-Thoracic Surgery, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Techonology, Tampere University, Tampere 33014, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Arvo Ylpön katu 34, Tampere 33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Techonology, Tampere University, Tampere 33014, Finland
| | - Anke H. Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, the Netherlands
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Departments of Medicine and Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Colin N. Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Division of Molecular and Clinical Medicine, Level 5, Mailbox 12, Ninewells Hospital and Medical School, Dundee, UK
| | - Adriaan O. Kraaijeveld
- Department of Cardiology, Division Heart and Lungs, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carl J. Pepine
- College of Medicine, Division of Cardiovascular Medicine, University of Florida, 1600 SW Archer Road/Box 100277, Gainesville, FL 32610, USA
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str, 31-066 Kraków, Poland
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Markus Scholz
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Tabassome Simon
- Assistance Publique-Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, Platform of Clinical Research of East Paris (URCEST-CRCEST-CRB HUEP-UPMC), FACT (French Alliance for Cardiovascular trials); Sorbonne Université, Paris-06, France
- Paris-Sorbonne University, UPMC-Site St Antoine, 27 Rue Chaligny, 75012, Paris, France
| | - John A. Spertus
- University of Missouri-Kansas City, Kansas City, Missouri, USA
- Saint Luke’s Mid America Heart Institute, 4401 Wornall Road, 9th Floor, Kansas City, MO 64111, USA
| | - Alexandre F. R. Stewart
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada
| | - Wojciech Szczeklik
- Department of Internal Medicine, Jagiellonian University Medical College, 8 Skawinska Str, 31-066 Kraków, Poland
| | - Joachim Thiery
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - Frank L.J. Visseren
- Department of Vascular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | | | - A. Mark Richards
- The Christchurch Heart Institute, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
- Cardiovascular Research Institute, National University of Singapore, 1 E Kent Ridge Road, Singapore
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Vicky A. Cameron
- The Christchurch Heart Institute, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
| | - Axel Åkerblom
- Uppsala Clinical Research Center, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Guillaume Pare
- McMaster University, Department of Pathology and Molecular Medicine, Hamilton, Canada
- Population Health Research Institute, Hamilton, ON L8L 2X2, Canada
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, LE3 9QP, UK
| | - Aroon D. Hingorani
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
| | - Jurriën M. ten Berg
- St. Antonius Hospital, department of Cardiology, Koekoekslaan 1, 3435CM, Nieuwegein, the Netherlands
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart and Lungs, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Riyaz Patel
- Institute of Cardiovascular Science and UCL BHF Research Accelerator, Faculty of Population Health Science, University College London, London, UK
- Bart’s Heart Centre, St Bartholomew’s Hospital, London, EC1A2DA, UK
| |
Collapse
|
10
|
Sipilä K, Tikkakoski A, Alanko S, Haarala A, Hernesniemi J, Lyytikäinen LP, Viik J, Lehtimäki T, Nieminen T, Nikus K, Kähönen M. Combination of low blood pressure response, low exercise capacity and slow heart rate recovery during an exercise test significantly increases mortality risk. Ann Med 2019; 51:390-396. [PMID: 31638839 PMCID: PMC7877875 DOI: 10.1080/07853890.2019.1684550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aims: We investigated the combination of low systolic blood pressure (SBP) response, low exercise capacity (EC) and slow heart rate recovery (HRR) during an exercise test in mortality prediction.Patients and methods: Our population consisted of 3456 patients from the Finnish Cardiovascular Study. A failure of SBP to increase >42 mmHg was defined as a low response. Low EC was defined as < 8 metabolic equivalents. 1-minute HRR ≤18 bpm from maximum was defined as slow HRR.Results: During a median follow up of 10.0 years, 537 participants died. Reduced SBP response, low EC and slow HRR were independent predictors of all-cause and CV mortality (p < .001 for all). Patients with reduced SBP response, low EC and slow HRR had a very high mortality rate of 42.1% during follow up compared to only 4.5% of the patients without any of these risk factors. The hazard ratios for all-cause mortality in patients with one, two or three of the studied risk factors were 3.2, 6.0, and 10.6, respectively (p < .001 for all).Conclusion: The combination of reduced SBP response, low exercise capacity, and reduced HRR in an exercise test is associated with very high mortality and can be used in risk stratification.Key messagesThe combination of low blood pressure response, low exercise capacity and slow heart rate recovery in an exercise test is able to identify a group of patients in a very high mortality risk.These parameters are easily derived from an exercise test.All parameters are commonly available in clinical practice.
Collapse
Affiliation(s)
- Kalle Sipilä
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland
| | - Antti Tikkakoski
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland
| | - Sanni Alanko
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland
| | - Atte Haarala
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland
| | - Jussi Hernesniemi
- Heart Center, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Jari Viik
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Terho Lehtimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Tuomo Nieminen
- Department of Internal Medicine, University of Helsinki, Helsinki University Central Hospital, Helsinki, Finland.,South Karelia Central Hospital, Lappeenranta, Finland
| | - Kjell Nikus
- Heart Center, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
11
|
Hernesniemi JA, Sipilä K, Tikkakoski A, Tynkkynen JT, Mishra PP, Lyytikäinen LP, Nikus K, Nieminen T, Lehtimaki T, Kähönen M. Cardiorespiratory fitness and heart rate recovery predict sudden cardiac death independent of ejection fraction. Heart 2019; 106:434-440. [PMID: 31422363 DOI: 10.1136/heartjnl-2019-315198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To evaluate whether cardiorespiratory fitness (CRF) and heart rate recovery (HRR) associate with the risk of sudden cardiac death (SCD) independently of left ventricular ejection fraction (LVEF). METHODS The Finnish Cardiovascular Study is a prospective clinical study of patients referred to clinical exercise testing in 2001-2008 and follow-up until December 2013. Patients without pacemakers undergoing first maximal or submaximal exercise testing with cycle ergometer were included (n=3776). CRF in metabolic equivalents (METs) was estimated by achieving maximal work level. HRR was defined as the reduction in heart rate 1 min after maximal exertion. Adjudication of SCD was based on death certificates. LVEF was measured for clinical indications in 71.4% of the patients (n=2697). RESULTS Population mean age was 55.7 years (SD 13.1; 61% men). 98 SCDs were recorded during a median follow-up of 9.1 years (6.9-10.7). Mean CRF and HRR were 7.7 (SD 2.9) METs and 25 (SD 12) beats/min/min. Both CRF and HRR were associated with the risk of SCD in the entire study population (HRCRF0.47 (0.37-0.59), p<0.001 and HRHRR0.57 (0.48-0.67), p<0.001 with HR estimates corresponding to one SD increase in the exposure variables) and with CRF, HRR and LVEF in the same model (HRCRF0.60 (0.45-0.79), p<0.001, HRHRR0.65 (0.51-0.82), p<0.001) or adjusting additionally for all significant risk factors for SCD (LVEF, sex, creatinine level, history of myocardial infarction and atrial fibrillation, corrected QT interval) (HRCRF0.69 (0.52-0.93), p<0.01, HRHRR0.74 (0.58-0.95) p=0.02). CONCLUSIONS CRF and HRR are significantly associated with the risk of SCD regardless of LVEF.
Collapse
Affiliation(s)
- Jussi A Hernesniemi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland .,Department of Cardiology, Tays Heart Hospital, Tampere University Hopsital, Tampere, Finland.,Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
| | - Kalle Sipilä
- Department of Clinical Physiology, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Antti Tikkakoski
- Department of Clinical Physiology, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Juho T Tynkkynen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Cardiology, Tays Heart Hospital, Tampere University Hopsital, Tampere, Finland.,Department of Radiology, Kanta-Häme Central Hospital, Hämeenlinna, Finland
| | - Pashupati P Mishra
- Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Cardiology, Tays Heart Hospital, Tampere University Hopsital, Tampere, Finland.,Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Kjell Nikus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Cardiology, Tays Heart Hospital, Tampere University Hopsital, Tampere, Finland.,Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
| | - Tuomo Nieminen
- Department of Internal Medicine, Päijät-Häme Central-Hospital, Lahti, Finland
| | - Terho Lehtimaki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Mika Kähönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Cardiology, Tays Heart Hospital, Tampere University Hopsital, Tampere, Finland.,Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland.,Department of Clinical Physiology, Tampere University Hospital, University of Tampere, Tampere, Finland
| |
Collapse
|
12
|
Rizas KD, Doller AJ, Hamm W, Vdovin N, von Stuelpnagel L, Zuern CS, Bauer A. Periodic repolarization dynamics as a risk predictor after myocardial infarction: Prospective validation study. Heart Rhythm 2019; 16:1223-1231. [PMID: 30818092 DOI: 10.1016/j.hrthm.2019.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Periodic repolarization dynamics (PRD) is a novel electrocardiographic phenomenon that refers to sympathetic activity-associated low-frequency modulations of cardiac repolarization. Retrospective post-myocardial infarction (MI) studies revealed that increased PRD indicates an increased risk of subsequent death. OBJECTIVE This is the first prospective study to validate PRD in patients after MI receiving up-to-date treatment. METHODS Four hundred fifty-five survivors of MI (age ≤80 years) in sinus rhythm were enrolled. PRD was assessed from 20-minute electrocardiographic recordings (2048 Hz) and prospectively dichotomized at 5.75 deg2. Primary and secondary end points were total mortality and cardiovascular mortality, respectively. Multivariable analyses additionally included Global Registry of Acute Coronary Events score (dichotomized at >140), left ventricular ejection fraction (dichotomized at ≤35%), diabetes mellitus, and deceleration capacity of heart rate (dichotomized at ≤2.5 ms). The prognostic power of PRD was evaluated using receiver operating characteristic curve analysis, Cox regression analysis, and the integrated discrimination improvement index. RESULTS During a median follow-up period of 27 months, 47 patients died. Twenty-three of these deaths were classified as cardiovascular. Increased PRD was significantly associated with both end points, yielding areas under receiver operating characteristic curves of 69.3% (60.2%-77.8%) and 79.1% (69.7%-86.7%) for total mortality and cardiovascular mortality, respectively (P < .001 for both). In multivariable analysis, increased PRD indicated a 2.2- and 9.5-fold risk of total mortality and cardiovascular mortality (P = .024 and P = .003, respectively). Addition of PRD to the models significantly improved the integrated discrimination improvement index for total (P = .047) and cardiovascular mortality (P = .007). CONCLUSION PRD is a strong and independent predictor of total mortality and cardiovascular mortality in patients after MI treated with contemporary therapy.
Collapse
Affiliation(s)
- Konstantinos D Rizas
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Angela J Doller
- Deutsches Herzkompetenz Zentrum, Abteilung Kardiologie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Wolfgang Hamm
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Nikolay Vdovin
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Lukas von Stuelpnagel
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Christine S Zuern
- Deutsches Herzkompetenz Zentrum, Abteilung Kardiologie, Universitätsklinikum Tübingen, Tübingen, Germany; Department of Cardiology, University Hospital, Basel, Switzerland
| | - Axel Bauer
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludwig-Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
13
|
Salokari E, Laukkanen JA, Lehtimaki T, Kurl S, Kunutsor S, Zaccardi F, Viik J, Lehtinen R, Nikus K, Kööbi T, Turjanmaa V, Kähönen M, Nieminen T. The Duke treadmill score with bicycle ergometer: Exercise capacity is the most important predictor of cardiovascular mortality. Eur J Prev Cardiol 2018; 26:199-207. [PMID: 30354741 PMCID: PMC6330693 DOI: 10.1177/2047487318804618] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background The Duke treadmill score, a widely used treadmill testing tool, is a weighted index combining exercise time or capacity, maximum ST-segment deviation and exercise-induced angina. No previous studies have investigated whether the Duke treadmill score and its individual components based on bicycle exercise testing predict cardiovascular death. Design Two populations with a standard bicycle testing were used: 3936 patients referred for exercise testing (2371 men, age 56 ± 13 years) from the Finnish Cardiovascular Study (FINCAVAS) and a population-based sample of 2683 men (age 53 ± 5.1 years) from the Kuopio Ischaemic Heart Disease study (KIHD). Methods Cox regression was applied for risk prediction with cardiovascular mortality as the primary endpoint. Results In FINCAVAS, during a median 6.3-year (interquartile range (IQR) 4.5–8.2) follow-up period, 180 patients (4.6%) experienced cardiovascular mortality. In KIHD, 562 patients (21.0%) died from cardiovascular causes during the median follow-up of 24.1 (IQR 18.0–26.2) years. The Duke treadmill score was associated with cardiovascular mortality in both populations (FINCAVAS, adjusted hazard ratio (HR) 3.15 for highest vs. lowest Duke treadmill score tertile, 95% confidence interval (CI) 1.83–5.42, P < 0.001; KIHD, adjusted HR 1.71, 95% CI 1.34–2.18, P < 0.001). However, after progressive adjustment for the Duke treadmill score components, the score was not associated with cardiovascular mortality in either study population, as exercise capacity in metabolic equivalents of task was the dominant harbinger of poor prognosis. Conclusions The Duke treadmill score is associated with cardiovascular mortality among patients who have undergone bicycle exercise testing, but metabolic equivalents of task, a component of the Duke treadmill score, proved to be a superior predictor.
Collapse
Affiliation(s)
- Esko Salokari
- 1 Department of Internal Medicine, University of Helsinki, Finland
| | - Jari A Laukkanen
- 2 Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Finland.,3 Department of Medicine, Central Finland Health Care District, Finland.,4 Faculty of Sport and Health Sciences, University of Jyväskylä, Finland
| | - Terho Lehtimaki
- 5 Department of Clinical Chemistry, University of Tampere, Finland
| | - Sudhir Kurl
- 2 Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Finland
| | - Setor Kunutsor
- 6 School of Clinical Sciences, University of Bristol, Southmead Hospital, Bristol, UK
| | | | - Jari Viik
- 8 Department of Biomedical Engineering, Tampere University of Technology, Finland
| | - Rami Lehtinen
- 9 Department of ICT and Health Technology, Tampere University of Applied Sciences, Finland
| | - Kjell Nikus
- 10 Faculty of Medicine and Life Sciences, University of Tampere and Heart Center, Finland
| | - Tiit Kööbi
- 11 Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Finland
| | | | - Mika Kähönen
- 11 Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Finland
| | - Tuomo Nieminen
- 13 Department of Internal Medicine, University of Helsinki, Finland.,14 South Karelia Central Hospital, Lappeenranta, Finland
| |
Collapse
|
14
|
Rinne P, Lyytikäinen LP, Raitoharju E, Kadiri JJ, Kholova I, Kähönen M, Lehtimäki T, Oksala N. Pro-opiomelanocortin and its Processing Enzymes Associate with Plaque Stability in Human Atherosclerosis - Tampere Vascular Study. Sci Rep 2018; 8:15078. [PMID: 30305673 PMCID: PMC6180013 DOI: 10.1038/s41598-018-33523-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/27/2018] [Indexed: 02/08/2023] Open
Abstract
α-melanocyte-stimulating hormone (α-MSH) is processed from pro-opiomelanocortin (POMC) and mediates anti-inflammatory actions in leukocytes. α-MSH also promotes macrophage reverse cholesterol transport by inducing ATP-binding cassette transporters ABCA1 and ABCG1. Here we investigated the regulation of POMC and α-MSH expression in atherosclerosis. First, transcript levels of POMC and its processing enzymes were analyzed in human arterial plaques (n = 68) and non-atherosclerotic controls (n = 24) as well as in whole blood samples from coronary artery disease patients (n = 55) and controls (n = 45) by microarray. POMC expression was increased in femoral plaques compared to control samples as well as in unstable advanced plaques. α-MSH-producing enzyme, carboxypeptidase E, was down-regulated, whereas prolylcarboxypeptidase, an enzyme inactivating α-MSH, was up-regulated in unstable plaques compared to stable plaques, suggesting a possible reduction in intraplaque α-MSH levels. Second, immunohistochemical analyses revealed the presence of α-MSH in atherosclerotic plaques and its localization in macrophages and other cell types. Lastly, supporting the role of α-MSH in reverse cholesterol transport, POMC expression correlated with ABCA1 and ABCG1 in human plaque and whole blood samples. In conclusion, α-MSH is expressed in atherosclerotic plaques and its processing enzymes associate with plaque stability, suggesting that measures to enhance the local bioavailability of α-MSH might protect against atherosclerosis.
Collapse
Affiliation(s)
- Petteri Rinne
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - James J Kadiri
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ivana Kholova
- Department of Pathology, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Niku Oksala
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland. .,Department of Surgery, Tampere University Hospital, Tampere, Finland and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland and Finnish Cardiovascular Research Center-Tampere, Tampere, Finland.
| |
Collapse
|
15
|
Rizas KD, McNitt S, Hamm W, Massberg S, Kääb S, Zareba W, Couderc JP, Bauer A. Prediction of sudden and non-sudden cardiac death in post-infarction patients with reduced left ventricular ejection fraction by periodic repolarization dynamics: MADIT-II substudy. Eur Heart J 2018; 38:2110-2118. [PMID: 28431133 PMCID: PMC5837472 DOI: 10.1093/eurheartj/ehx161] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
Aims To test the value of Periodic Repolarization Dynamics (PRD), a recently validated electrocardiographic marker of sympathetic activity, as a novel approach to predict sudden cardiac death (SCD) and non-sudden cardiac death (N-SCD) and to improve identification of patients that profit from ICD-implantation. Methods and results We included 856 post-infarction patients with left-ventricular ejection fraction (LVEF) ≤30% of the MADIT-II trial in sinus rhythm. Of these, 507 and 348 patients were randomized to ICD or conventional treatment. PRD was assessed from multipolar 10-min baseline ECGs. Primary and secondary endpoints were total mortality, SCD and N-SCD. Multivariable analyses included treatment group, QRS-duration, New York Heart Association classification, blood-urea nitrogen, diabetes mellitus, beta-blocker therapy and LVEF. During follow-up of 20.4 months, 119 patients died (53 SCD and 36 N-SCD). On multivariable analyses, increased PRD was a significant predictor of mortality (standardized coefficient 1.37[1.19–1.59]; P < 0.001) and SCD (1.40 [1.13–1.75]; P = 0.003) but also predicted N-SCD (1.41[1.10–1.81]; P = 0.006). While increased PRD predicted SCD in conventionally treated patients (1.61[1.23–2.11]; P < 0.001), it was predictive of N-SCD (1.63[1.28–2.09]; P < 0.001) and adequate ICD-therapies (1.20[1.03–1.39]; P = 0.017) in ICD-treated patients. ICD-treatment substantially reduced mortality in the lowest three PRD-quartiles by 53% (P = 0.001). However, there was no effect in the highest PRD-quartile (mortality increase by 29%; P = 0.412; P < 0.001 for difference) as the reduction of SCD was compensated by an increase of N-SCD. Conclusion In post-infarction patients with impaired LVEF, PRD is a significant predictor of SCD and N-SCD. Assessment of PRD is a promising tool to identify post-MI patients with reduced LVEF who might benefit from intensified treatment.
Collapse
Affiliation(s)
- Konstantinos D Rizas
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Biedersteiner Str. 29, 80802 Munich, Germany.,Abteilung Kardiologie, Deutsches Herzkompetenz Zentrum, Universitätsklinikum Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen
| | - Scott McNitt
- Heart Research Follow-Up Program, University of Rochester Medical Center, 265 Crittenden Blvd, Rochester, NY 14642, USA
| | - Wolfgang Hamm
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Wojciech Zareba
- Heart Research Follow-Up Program, University of Rochester Medical Center, 265 Crittenden Blvd, Rochester, NY 14642, USA
| | - Jean-Philippe Couderc
- Heart Research Follow-Up Program, University of Rochester Medical Center, 265 Crittenden Blvd, Rochester, NY 14642, USA
| | - Axel Bauer
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Biedersteiner Str. 29, 80802 Munich, Germany.,Abteilung Kardiologie, Deutsches Herzkompetenz Zentrum, Universitätsklinikum Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen
| |
Collapse
|
16
|
Genetic Interactions with Age, Sex, Body Mass Index, and Hypertension in Relation to Atrial Fibrillation: The AFGen Consortium. Sci Rep 2017; 7:11303. [PMID: 28900195 PMCID: PMC5595875 DOI: 10.1038/s41598-017-09396-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022] Open
Abstract
It is unclear whether genetic markers interact with risk factors to influence atrial fibrillation (AF) risk. We performed genome-wide interaction analyses between genetic variants and age, sex, hypertension, and body mass index in the AFGen Consortium. Study-specific results were combined using meta-analysis (88,383 individuals of European descent, including 7,292 with AF). Variants with nominal interaction associations in the discovery analysis were tested for association in four independent studies (131,441 individuals, including 5,722 with AF). In the discovery analysis, the AF risk associated with the minor rs6817105 allele (at the PITX2 locus) was greater among subjects ≤ 65 years of age than among those > 65 years (interaction p-value = 4.0 × 10−5). The interaction p-value exceeded genome-wide significance in combined discovery and replication analyses (interaction p-value = 1.7 × 10−8). We observed one genome-wide significant interaction with body mass index and several suggestive interactions with age, sex, and body mass index in the discovery analysis. However, none was replicated in the independent sample. Our findings suggest that the pathogenesis of AF may differ according to age in individuals of European descent, but we did not observe evidence of statistically significant genetic interactions with sex, body mass index, or hypertension on AF risk.
Collapse
|
17
|
von Essen M, Rahikainen R, Oksala N, Raitoharju E, Seppälä I, Mennander A, Sioris T, Kholová I, Klopp N, Illig T, Karhunen PJ, Kähönen M, Lehtimäki T, Hytönen VP. Talin and vinculin are downregulated in atherosclerotic plaque; Tampere Vascular Study. Atherosclerosis 2016; 255:43-53. [PMID: 27816808 DOI: 10.1016/j.atherosclerosis.2016.10.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS Focal adhesions (FA) play an important role in the tissue remodeling and in the maintenance of tissue integrity and homeostasis. Talin and vinculin proteins are among the major constituents of FAs contributing to cellular well-being and intercellular communication. METHODS Microarray analysis (MA) and qRT-PCR low-density array were implemented to analyze talin-1, talin-2, meta-vinculin and vinculin gene expression in circulating blood and arterial plaque. RESULTS All analyzed genes were significantly and consistently downregulated in plaques (carotid, abdominal aortic and femoral regions) compared to left internal thoracic artery (LITA) control. The use of LITA samples as controls for arterial plaque samples was validated using immunohistochemistry by comparing LITA samples with healthy arterial samples from a cadaver. Even though the differences in expression levels between stable and unstable plaques were not statistically significant, we observed further negative tendency in the expression in unstable atherosclerotic plaques. The confocal tissue imaging revealed gradient of talin-1 expression in plaque with reduction close to the vessel lumen. Similar gradient was observed for talin-2 expression in LITA controls but was not detected in plaques. This suggests that impaired tissue mechanostability affects the tissue remodeling and healing capabilities leading to development of unstable plaques. CONCLUSIONS The central role of talin and vinculin in cell adhesions suggests that the disintegration of the tissue in atherosclerosis could be partially driven by downregulation of these genes, leading to loosening of cell-ECM interactions and remodeling of the tissue.
Collapse
Affiliation(s)
- Magdaléna von Essen
- BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
| | - Rolle Rahikainen
- BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
| | - Niku Oksala
- Dep. of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland; Division of Vascular Surgery, Department of Surgery, Tampere University Hospital, Tampere, Finland
| | - Emma Raitoharju
- Dep. of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - Ilkka Seppälä
- Dep. of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - Ari Mennander
- Heart Center, Tampere University Hospital, Tampere, Finland
| | - Thanos Sioris
- Heart Center, Tampere University Hospital, Tampere, Finland
| | - Ivana Kholová
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - Norman Klopp
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany; Institute of Human Genetics, Hannover Medical School, Hannover, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Pekka J Karhunen
- School of Medicine, University of Tampere and Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - Terho Lehtimäki
- Dep. of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - Vesa P Hytönen
- BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland.
| |
Collapse
|
18
|
Oksala N, Pärssinen J, Seppälä I, Klopp N, Illig T, Laaksonen R, Levula M, Raitoharju E, Kholova I, Sioris T, Kähönen M, Lehtimäki T, Hytönen VP. Kindlin 3 (FERMT3) is associated with unstable atherosclerotic plaques, anti-inflammatory type II macrophages and upregulation of beta-2 integrins in all major arterial beds. Atherosclerosis 2015; 242:145-54. [PMID: 26188538 DOI: 10.1016/j.atherosclerosis.2015.06.058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/27/2015] [Accepted: 06/29/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Kindlins (FERMT) are cytoplasmic proteins required for integrin (ITG) activation, leukocyte transmigration, platelet aggregation and thrombosis. Characterization of kindlins and their association with atherosclerotic plaques in human(s) is lacking. METHODS AND RESULTS Exploratory microarray (MA) was first performed followed by selective quantitative validation of robustly expressed genes with qRT-PCR low-density array (LDA). In LDA, ITGA1 (1.30-fold, p = 0.041) and ITGB3 (1.37-fold, p = 0.036) were upregulated in whole blood samples of patients with coronary artery disease (CAD) compared to healthy controls. In arterial plaques, both robustly expressed transcript variants of FERMT3 (MA: 5.90- and 3.4-fold; LDA: 3.99-fold, p < 0.0001 for all) and ITGB2 (MA: 4.81- and 4.92-fold; LDA: 5.29-fold, p < 0.0001 for all) were upregulated while FERMT2 was downregulated (MA: -1.61-fold; LDA: -2.88-fold, p < 0.0001 for both). The other integrins (ITGA1, ITGAV, ITGB3, ITGB5) were downregulated. All these results were replicated in at least one arterial bed. The latter FERMT3 transcript variant associated with unstable plaques (p = 0.0004). FERMT3 correlated with M2 macrophage markers and in hierarchical cluster analysis clustered with inflammatory and macrophage markers, while FERMT2 correlated with SMC-rich plaque markers and clustered with SMC markers. In confocal immunofluorescence analysis, FERMT3 protein colocalized with abundant CD68-positive cells of monocytic origin in the atherosclerotic plaques, while co-localization of FERMT3 with HHF35 indicative of smooth muscle cells was low. CONCLUSIONS Kindlin-3 (FERMT3) is upregulated in atherosclerotic, especially unstable plaques, mainly in cells of monocytic origin and of M2 type. Simultaneous upregulation of ITGB2 suggests a synergistic effect on leukocyte adherence and transmigration into the vessel wall.
Collapse
Affiliation(s)
- Niku Oksala
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland; School of Medicine, University of Tampere, Finland; Division of Vascular Surgery, Department of Surgery, Tampere University Hospital, Finland.
| | - Jenita Pärssinen
- BioMediTech, University of Tampere, Tampere, Finland and Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Norman Klopp
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum, German Research Center for Environmental Health, Munich, Germany; Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum, German Research Center for Environmental Health, Munich, Germany; Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Reijo Laaksonen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland; School of Medicine, University of Tampere, Finland
| | - Mari Levula
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Ivana Kholova
- Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Thanos Sioris
- Heart Center, Tampere University Hospital, Tampere, Finland
| | - Mika Kähönen
- School of Medicine, University of Tampere, Finland; Division of Vascular Surgery, Department of Surgery, Tampere University Hospital, Finland; Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland; School of Medicine, University of Tampere, Finland
| | - Vesa P Hytönen
- BioMediTech, University of Tampere, Tampere, Finland and Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
19
|
Zimmermann E, Ängquist LH, Mirza SS, Zhao JH, Chasman DI, Fischer K, Qi Q, Smith AV, Thinggaard M, Jarczok MN, Nalls MA, Trompet S, Timpson NJ, Schmidt B, Jackson AU, Lyytikäinen LP, Verweij N, Mueller-Nurasyid M, Vikström M, Marques-Vidal P, Wong A, Meidtner K, Middelberg RP, Strawbridge RJ, Christiansen L, Kyvik KO, Hamsten A, Jääskeläinen T, Tjønneland A, Eriksson JG, Whitfield JB, Boeing H, Hardy R, Vollenweider P, Leander K, Peters A, van der Harst P, Kumari M, Lehtimäki T, Meirhaeghe A, Tuomilehto J, Jöckel KH, Ben-Shlomo Y, Sattar N, Baumeister SE, Smith GD, Casas JP, Houston DK, März W, Christensen K, Gudnason V, Hu FB, Metspalu A, Ridker PM, Wareham NJ, Loos RJF, Tiemeier H, Sonestedt E, Sørensen TIA. Is the adiposity-associated FTO gene variant related to all-cause mortality independent of adiposity? Meta-analysis of data from 169,551 Caucasian adults. Obes Rev 2015; 16:327-340. [PMID: 25752329 PMCID: PMC4564522 DOI: 10.1111/obr.12263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/12/2014] [Indexed: 11/28/2022]
Abstract
Previously, a single nucleotide polymorphism (SNP), rs9939609, in the FTO gene showed a much stronger association with all-cause mortality than expected from its association with body mass index (BMI), body fat mass index (FMI) and waist circumference (WC). This finding implies that the SNP has strong pleiotropic effects on adiposity and adiposity-independent pathological pathways that leads to increased mortality. To investigate this further, we conducted a meta-analysis of similar data from 34 longitudinal studies including 169,551 adult Caucasians among whom 27,100 died during follow-up. Linear regression showed that the minor allele of the FTO SNP was associated with greater BMI (n = 169,551; 0.32 kg m(-2) ; 95% CI 0.28-0.32, P < 1 × 10(-32) ), WC (n = 152,631; 0.76 cm; 0.68-0.84, P < 1 × 10(-32) ) and FMI (n = 48,192; 0.17 kg m(-2) ; 0.13-0.22, P = 1.0 × 10(-13) ). Cox proportional hazard regression analyses for mortality showed that the hazards ratio (HR) for the minor allele of the FTO SNPs was 1.02 (1.00-1.04, P = 0.097), but the apparent excess risk was eliminated after adjustment for BMI and WC (HR: 1.00; 0.98-1.03, P = 0.662) and for FMI (HR: 1.00; 0.96-1.04, P = 0.932). In conclusion, this study does not support that the FTO SNP is associated with all-cause mortality independently of the adiposity phenotypes.
Collapse
Affiliation(s)
- E Zimmermann
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Copenhagen, Denmark
| | - L H Ängquist
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Copenhagen, Denmark
| | - S S Mirza
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - J H Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - D I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - K Fischer
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Q Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, New York, USA
| | - A V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Icelandic, Reykajvik, Iceland
| | - M Thinggaard
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - M N Jarczok
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M A Nalls
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - S Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - N J Timpson
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - B Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany
| | - A U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - L P Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- School of Medicine, University of Tampere, Tampere, Finland
| | - N Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Mueller-Nurasyid
- Department of Medicine I, Ludwig-Maximilians-University, Munich, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Deutsches Forschungszentrum für Herz-Kreislauferkrankungen (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - M Vikström
- Karolinska Institutet, Institute of Environmental Medicine, Unit of Cardiovascular Epidemiology, Stockholm, Sweden
| | - P Marques-Vidal
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - A Wong
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - K Meidtner
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - R P Middelberg
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - R J Strawbridge
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - L Christiansen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - K O Kyvik
- Institute of Regional Health Services Research and Odense Patient data Explorative Network, Odense University Hospital, Odense, Denmark
| | - A Hamsten
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - T Jääskeläinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - A Tjønneland
- Danish Cancer Society Research Centre, Copenhagen, Denmark
| | - J G Eriksson
- Diabetes Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
| | - J B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - H Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - R Hardy
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - P Vollenweider
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - K Leander
- Karolinska Institutet, Institute of Environmental Medicine, Unit of Cardiovascular Epidemiology, Stockholm, Sweden
| | - A Peters
- Deutsches Forschungszentrum für Herz-Kreislauferkrankungen (DZHK), Partner site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - P van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Genetic, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Neterlands Heart Institute, Utrecht, The Netherlands
| | - M Kumari
- Department of Epidemiology and Public Health, University College London, London, UK
- ISER, University of Essex, Colchester, UK
| | - T Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- School of Medicine, University of Tampere, Tampere, Finland
| | - A Meirhaeghe
- Inserm, U744, Institut Pasteur de Lille, University Lille Nord de France, Lille, France
| | - J Tuomilehto
- Diabetes Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
- Centre for Vascular Prevention, Danube-University Krems, Krems, Austria
- Instituto de Investigacion Sanitaria del Hospital Universario LaPaz (IdiPAZ), Madrid, Spain
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - K-H Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany
| | - Y Ben-Shlomo
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - N Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, UK
| | - S E Baumeister
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - G Davey Smith
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - J P Casas
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - D K Houston
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - W März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany
| | - K Christensen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - V Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Icelandic, Reykajvik, Iceland
| | - F B Hu
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - A Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - P M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - N J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - R J F Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - H Tiemeier
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - E Sonestedt
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - T I A Sørensen
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section on Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
20
|
Turpeinen H, Seppälä I, Lyytikäinen LP, Raitoharju E, Hutri-Kähönen N, Levula M, Oksala N, Waldenberger M, Klopp N, Illig T, Mononen N, Laaksonen R, Raitakari O, Kähönen M, Lehtimäki T, Pesu M. A genome-wide expression quantitative trait loci analysis of proprotein convertase subtilisin/kexin enzymes identifies a novel regulatory gene variant for FURIN expression and blood pressure. Hum Genet 2015; 134:627-36. [PMID: 25813623 DOI: 10.1007/s00439-015-1546-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/18/2015] [Indexed: 01/11/2023]
Abstract
Proprotein convertase subtilisin/kexin (PCSK) enzymes cleave and convert their immature substrates into biologically active forms. Polymorphisms in the PCSK genes have been reported to associate with human diseases and phenotypes, including hypercholesterolemia and blood pressure (BP), and targeting PCSKs is considered a promising future form of drug therapy. PCSK processing is readily induced upon upregulation of the enzyme, but the genetic factors contributing to PCSK expression have not been thoroughly characterized. To gain a comprehensive understanding of the genetic regulation of PCSK expression, we performed, for the first time, a genome-wide expression quantitative trait loci (eQTL) analysis using mRNA expression in >1400 human peripheral blood samples from the Cardiovascular Risk in Young Finns Study and ca. ten million single-nucleotide polymorphisms (SNPs). The expression data showed clear expression for FURIN, PCSK5, PCSK7 and MBTPS1 (membrane-bound transcription factor peptidase, site 1) mRNAs in virtually all tested samples. A discovery analysis demonstrated a genome-wide significant (p < 5 × 10(-8)) association with the selected PCSK probes for 1024 variants, which were located at ten independent loci. Of these loci, 5/10 could be confirmed to regulate PCSK expression in two additional and independent sample sets. Finally, a phenotypic analysis demonstrated that a novel cis-eQTL SNP rs4702 for FURIN is strongly associated with both diastolic (p = 0.012) and systolic (p = 0.035) BP levels, as well as peripheral vascular resistance (p = 0.003). These findings indicate that the expression of the PCSK enzymes is regulated by genetic factors, which have biological roles in health and disease.
Collapse
Affiliation(s)
- Hannu Turpeinen
- BioMediTech, University of Tampere, Biokatu 8, 33580, Tampere, Finland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ghisi GLDM, Grace SL, Thomas S, Evans MF, Oh P. Development and psychometric validation of the second version of the Coronary Artery Disease Education Questionnaire (CADE-Q II). PATIENT EDUCATION AND COUNSELING 2015; 98:378-383. [PMID: 25481574 DOI: 10.1016/j.pec.2014.11.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/10/2014] [Accepted: 11/17/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVES To develop and psychometrically-validate a revised version of the Coronary Artery Disease Education Questionnaire (CADE-Q)--a tool to assess patients' knowledge about CAD in cardiac rehabilitation (CR). METHODS After a needs assessment, a literature review and focus group with CR experts, the revised questionnaire was developed. It underwent pilot-testing in 30 patients, which lead to further refinement. The questionnaire was then psychometrically-tested in 307 CR patients. Internal consistency was assessed using Cronbach's alpha, the dimensional structure through exploratory factor analysis, and criterion validity with regard to educational level. RESULTS Cronbach's alpha was 0.91. Criterion validity was supported by significant differences in mean scores by educational level (p<0.001). Factor analysis revealed four factors, which were internally-consistent (0.65-0.77), and well-defined by items. The mean total score was 64.2±18.1/93. Patients with a history of heart failure, cardiomyopathy and percutaneous coronary intervention (p<0.05) had significantly higher knowledge scores compared with patients without such a history. Knowledge about exercise and their medical condition was significantly higher than risk factors, nutrition and psychosocial risk. CONCLUSIONS The CADE-QII has good reliability and validity. PRACTICAL IMPLICATIONS This tool may be useful to assess CR participants' knowledge gaps, and to evaluate the efficacy of educational delivery in CR.
Collapse
Affiliation(s)
- Gabriela Lima de Melo Ghisi
- Exercise Sciences Department, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Canada; Cardiac Rehabilitation and Prevention Program, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada.
| | - Sherry L Grace
- Cardiac Rehabilitation and Prevention Program, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada; School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Scott Thomas
- Exercise Sciences Department, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Canada
| | | | - Paul Oh
- Cardiac Rehabilitation and Prevention Program, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
| |
Collapse
|
22
|
Minkkinen M, Nieminen T, Verrier RL, Leino J, Lehtimäki T, Viik J, Lehtinen R, Nikus K, Kööbi T, Turjanmaa V, Kähönen M. Prognostic capacity of a clinically indicated exercise test for cardiovascular mortality is enhanced by combined analysis of exercise capacity, heart rate recovery and T-wave alternans. Eur J Prev Cardiol 2014; 22:1162-70. [DOI: 10.1177/2047487314557190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 10/07/2014] [Indexed: 11/15/2022]
Affiliation(s)
- Mikko Minkkinen
- Department of Internal Medicine, Helsinki University Central Hospital, Finland
- Department of Clinical Physiology, Tampere University Hospital, Finland
| | - Tuomo Nieminen
- Department of Internal Medicine, University of Helsinki and South Karelia Central Hospital, Lappeenranta, Finland
| | - Richard L Verrier
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Johanna Leino
- Department of Clinical Physiology, Tampere University Hospital, Finland
- School of Medicine, University of Tampere, Finland
| | - Terho Lehtimäki
- School of Medicine, University of Tampere, Finland
- Department of Clinical Chemistry, Tampere University Hospital, Finland
| | - Jari Viik
- Department of Biomedical Engineering, Tampere University of Technology and BioMeditech, Tampere, Finland
| | | | - Kjell Nikus
- Department of Cardiology, Tampere University Hospital, Finland
| | - Tiit Kööbi
- Department of Clinical Physiology, Tampere University Hospital, Finland
- School of Medicine, University of Tampere, Finland
| | | | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Finland
- School of Medicine, University of Tampere, Finland
| |
Collapse
|
23
|
Pradhapan P, Tarvainen MP, Nieminen T, Lehtinen R, Nikus K, Lehtimäki T, Kähönen M, Viik J. Effect of heart rate correction on pre- and post-exercise heart rate variability to predict risk of mortality-an experimental study on the FINCAVAS cohort. Front Physiol 2014; 5:208. [PMID: 24917825 PMCID: PMC4042064 DOI: 10.3389/fphys.2014.00208] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/14/2014] [Indexed: 11/13/2022] Open
Abstract
The non-linear inverse relationship between RR-intervals and heart rate (HR) contributes significantly to the heart rate variability (HRV) parameters and their performance in mortality prediction. To determine the level of influence HR exerts over HRV parameters' prognostic power, we studied the predictive performance for different HR levels by applying eight correction procedures, multiplying or dividing HRV parameters by the mean RR-interval (RRavg) to the power 0.5-16. Data collected from 1288 patients in The Finnish Cardiovascular Study (FINCAVAS), who satisfied the inclusion criteria, was used for the analyses. HRV parameters (RMSSD, VLF Power and LF Power) were calculated from 2-min segment in the rest phase before exercise and 2-min recovery period immediately after peak exercise. Area under the receiver operating characteristic curve (AUC) was used to determine the predictive performance for each parameter with and without HR corrections in rest and recovery phases. The division of HRV parameters by segment's RRavg to the power 2 (HRVDIV-2) showed the highest predictive performance under the rest phase (RMSSD: 0.67/0.66; VLF Power: 0.70/0.62; LF Power: 0.79/0.65; cardiac mortality/non-cardiac mortality) with minimum correlation to HR (r = -0.15 to 0.15). In the recovery phase, Kaplan-Meier (KM) survival analysis revealed good risk stratification capacity at HRVDIV-2 in both groups (cardiac and non-cardiac mortality). Although higher powers of correction (HRVDIV-4and HRVDIV-8) improved predictive performance during recovery, they induced an increased positive correlation to HR. Thus, we inferred that predictive capacity of HRV during rest and recovery is augmented when its dependence on HR is weakened by applying appropriate correction procedures.
Collapse
Affiliation(s)
- Paruthi Pradhapan
- Department of Electronics and Communication Engineering, Tampere University of TechnologyTampere, Finland
- BioMediTechTampere, Finland
| | - Mika P. Tarvainen
- Department of Applied Physics, University of Eastern FinlandKuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University HospitalKuopio, Finland
| | - Tuomo Nieminen
- Heart and Lung Centre, Helsinki University Central HospitalHelsinki, Finland
| | - Rami Lehtinen
- Tampere Polytechnic, University of Applied SciencesTampere, Finland
| | - Kjell Nikus
- School of Medicine, University of TampereTampere, Finland
- Heart Centre, Department of Cardio-Thoracic Surgery, Tampere University HospitalTampere, Finland
| | - Terho Lehtimäki
- School of Medicine, University of TampereTampere, Finland
- Fimlab Laboratories, Department of Clinical ChemistryTampere, Finland
| | - Mika Kähönen
- School of Medicine, University of TampereTampere, Finland
- Department of Clinical Physiology, Tampere University HospitalTampere, Finland
| | - Jari Viik
- Department of Electronics and Communication Engineering, Tampere University of TechnologyTampere, Finland
- BioMediTechTampere, Finland
| |
Collapse
|
24
|
Rizas KD, Nieminen T, Barthel P, Zürn CS, Kähönen M, Viik J, Lehtimäki T, Nikus K, Eick C, Greiner TO, Wendel HP, Seizer P, Schreieck J, Gawaz M, Schmidt G, Bauer A. Sympathetic activity-associated periodic repolarization dynamics predict mortality following myocardial infarction. J Clin Invest 2014; 124:1770-80. [PMID: 24642467 DOI: 10.1172/jci70085] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/16/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Enhanced sympathetic activity at the ventricular myocardium can destabilize repolarization, increasing the risk of death. Sympathetic activity is known to cluster in low-frequency bursts; therefore, we hypothesized that sympathetic activity induces periodic low-frequency changes of repolarization. We developed a technique to assess the sympathetic effect on repolarization and identified periodic components in the low-frequency spectral range (≤0.1 Hz), which we termed periodic repolarization dynamics (PRD). METHODS We investigated the physiological properties of PRD in multiple experimental studies, including a swine model of steady-state ventilation (n=7) and human studies involving fixed atrial pacing (n=10), passive head-up tilt testing (n=11), low-intensity exercise testing (n=11), and beta blockade (n=10). We tested the prognostic power of PRD in 908 survivors of acute myocardial infarction (MI). Finally, we tested the predictive values of PRD and T-wave alternans (TWA) in 2,965 patients undergoing clinically indicated exercise testing. RESULTS PRD was not related to underlying respiratory activity (P<0.001) or heart-rate variability (P=0.002). Furthermore, PRD was enhanced by activation of the sympathetic nervous system, and pharmacological blockade of sympathetic nervous system activity suppressed PRD (P≤0.005 for both). Increased PRD was the strongest single risk predictor of 5-year total mortality (hazard ratio 4.75, 95% CI 2.94-7.66; P<0.001) after acute MI. In patients undergoing exercise testing, the predictive value of PRD was strong and complementary to that of TWA. CONCLUSION We have described and identified low-frequency rhythmic modulations of repolarization that are associated with sympathetic activity. Increased PRD can be used as a predictor of mortality in survivors of acute MI and patients undergoing exercise testing. TRIAL REGISTRATION ClinicalTrials.gov NCT00196274. FUNDING This study was funded by Angewandte Klinische Forschung, University of Tübingen (252-1-0).
Collapse
|
25
|
Oksala N, Pärssinen J, Seppälä I, Raitoharju E, Ivana K, Hernesniemi J, Lyytikäinen LP, Levula M, Mäkelä KM, Sioris T, Kähönen M, Laaksonen R, Hytönen V, Lehtimäki T. Association of Neuroimmune Guidance Cue Netrin-1 and Its Chemorepulsive Receptor UNC5B With Atherosclerotic Plaque Expression Signatures and Stability in Human(s). ACTA ACUST UNITED AC 2013; 6:579-87. [DOI: 10.1161/circgenetics.113.000141] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background—
Macrophage (MΦ) infiltration and smooth muscle cell (SMC) proliferation are hallmarks of atherosclerosis and unstable plaques. Neuroimmune guidance cue 1 (netrin-1 [NTN1]) plays a critical role controlling MΦ trafficking and SMC activation. Characterization of expression of NTN1 and its receptors and their association with plaque stability in human(s) is lacking.
Methods and Results—
The expression of NTN1 and its receptors did not differ in either whole blood or circulating monocytes from patients with coronary artery disease (n=55) compared with healthy controls (n=45). However, NTN1 was downregulated (−2.9-fold;
P
<0.0001) and UNC5B upregulated (2.2-fold;
P
<0.0001) in atherosclerotic plaques (n=68), whereas there were no differences in other NTN1 receptors compared with histologically normal controls (n=28). Increased UNC5B expression is associated with histologically more stable plaques (
P
=0.011). NTN1 expression correlated positively with SMC markers and signatures and negatively with inflammatory markers and M1 and especially M2 signatures in the atherosclerotic plaques. UNC5B clustering correlated positively with inflammatory and MΦ markers. NTN1 protein colocalized with CD68-positive cells of monocytic origin and muscle-actin-specific-antibody (HHF3)-positive cells indicative of SMCs in the plaques and only with SMCs in the control samples. NTN1 protein was highly expressed in the intimal layer of the control vessels.
Conclusions—
Present findings provide support for the hypothesis that dysregulation of expression of NTN1 in SMCs and its chemorepulsive receptor UNC5B in macrophages are involved in the development of atherosclerosis and unstable plaques.
Collapse
Affiliation(s)
- Niku Oksala
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jenita Pärssinen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Ilkka Seppälä
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Emma Raitoharju
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Kholova Ivana
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jussi Hernesniemi
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Mari Levula
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Kari-Matti Mäkelä
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Thanos Sioris
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Mika Kähönen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Reijo Laaksonen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Vesa Hytönen
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Terho Lehtimäki
- From the Department of Clinical Chemistry (N.O., I.S., E.R., J.H., L.-P.L., M.L., K.-M.M., R.L., T.L.), Institute of Biomedical Technology and BioMediTech (J.P., V.H.), and Department of Pathology (K.I.), Fimlab Laboratories, University of Tampere and Tampere University Hospital, Tampere, Finland; and Division of Vascular Surgery, Department of Surgery (N.O.) and Heart Center (T.S.), and Department of Clinical Physiology (M.K.), Tampere University Hospital, University of Tampere, Tampere, Finland
| |
Collapse
|
26
|
Kleber ME, Seppälä I, Pilz S, Hoffmann MM, Tomaschitz A, Oksala N, Raitoharju E, Lyytikäinen LP, Mäkelä KM, Laaksonen R, Kähönen M, Raitakari OT, Huang J, Kienreich K, Fahrleitner-Pammer A, Drechsler C, Krane V, Boehm BO, Koenig W, Wanner C, Lehtimäki T, März W, Meinitzer A. Genome-Wide Association Study Identifies 3 Genomic Loci Significantly Associated With Serum Levels of Homoarginine. ACTA ACUST UNITED AC 2013; 6:505-13. [DOI: 10.1161/circgenetics.113.000108] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background—
Low serum levels of the amino acid derivative, homoarginine, have been associated with increased risk of total and cardiovascular mortality. Homoarginine deficiency may be related to renal and heart diseases, but the pathophysiologic role of homoarginine and the genetic regulation of its serum levels are largely unknown.
Methods and Results—
In 3041 patients of the Ludwigshafen Risk and Cardiovascular Health (LURIC) study referred for coronary angiography and 2102 participants of the Young Finns Study (YFS), we performed a genome-wide association study to identify genomic loci associated with homoarginine serum levels and tested for associations of identified single-nucleotide polymorphisms with mortality in LURIC. We found genome-wide significant associations with homoarginine serum levels on chromosome 2 at the carbamoyl phosphate synthetase I locus, on chromosome 5 at the alanine-glyoxylate aminotransferase 2 locus, and on chromosome 15 at the glycine amidinotransferase locus, as well as a suggestive association on chromosome 6 at the Homo sapiens mediator complex subunit 23 gene/arginase I locus. All loci harbor enzymes located in the mitochondrium are involved in arginine metabolism. The strongest association was observed for rs1153858 at the glycine amidinotransferase locus with a
P
value of 1.25E-45 in the combined analysis and has been replicated in both the Die Deutsche Diabetes Dialyse Studie (4D study) and the Graz Endocrine Causes of Hypertension (GECOH) study.
Conclusions—
In our genome-wide association study, we identified 3 chromosomal regions significantly associated with serum homoarginine and another region with suggestive association, providing novel insights into the genetic regulation of homoarginine.
Collapse
|
27
|
Mäkelä KM, Seppälä I, Hernesniemi JA, Lyytikäinen LP, Oksala N, Kleber ME, Scharnagl H, Grammer TB, Baumert J, Thorand B, Jula A, Hutri-Kähönen N, Juonala M, Laitinen T, Laaksonen R, Karhunen PJ, Nikus KC, Nieminen T, Laurikka J, Kuukasjärvi P, Tarkka M, Viik J, Klopp N, Illig T, Kettunen J, Ahotupa M, Viikari JSA, Kähönen M, Raitakari OT, Karakas M, Koenig W, Boehm BO, Winkelmann BR, März W, Lehtimäki T. Genome-wide association study pinpoints a new functional apolipoprotein B variant influencing oxidized low-density lipoprotein levels but not cardiovascular events: AtheroRemo Consortium. ACTA ACUST UNITED AC 2012; 6:73-81. [PMID: 23247145 DOI: 10.1161/circgenetics.112.964965] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Oxidized low-density lipoprotein may be a key factor in the development of atherosclerosis. We performed a genome-wide association study on oxidized low-density lipoprotein and tested the impact of associated single-nucleotide polymorphisms (SNPs) on the risk factors of atherosclerosis and cardiovascular events. METHODS AND RESULTS A discovery genome-wide association study was performed on a population of young healthy white individuals (N=2080), and the SNPs associated with a P<5×10(-8) were replicated in 2 independent samples (A: N=2912; B: N=1326). Associations with cardiovascular endpoints were also assessed with 2 additional clinical cohorts (C: N=1118; and D: N=808). We found 328 SNPs associated with oxidized low-density lipoprotein. The genetic variant rs676210 (Pro2739Leu) in apolipoprotein B was the proxy SNP behind all associations (P=4.3×10(-136), effect size=13.2 U/L per allele). This association was replicated in the 2 independent samples (A and B, P=2.5×10(-47) and 1.1×10(-11), effect sizes=10.3 U/L and 7.8 U/L, respectively). In the meta-analyses of cohorts A, C, and D (excluding cohort B without angiographic data), the top SNP did not associate significantly with the age of onset of angiographically verified coronary artery disease (hazard ratio=1.00 [0.94-1.06] per allele), 3-vessel coronary artery disease (hazard ratio=1.03 [0.94-1.13]), or myocardial infarction (hazard ratio=1.04 [0.96-1.12]). CONCLUSIONS This novel genetic marker is an important factor regulating oxidized low-density lipoprotein levels but not a major genetic factor for the studied cardiovascular endpoints.
Collapse
Affiliation(s)
- Kari-Matti Mäkelä
- Department of Clinical Chemistry, Finn-Medi 2, PO Box 2000, FI-33521 Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Burattini L, Man S, Burattini R, Swenne CA. Comparison of standard versus orthogonal ECG leads for T-wave alternans identification. Ann Noninvasive Electrocardiol 2012; 17:130-40. [PMID: 22537331 DOI: 10.1111/j.1542-474x.2012.00490.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
T-wave alternans (TWA), an electrophysiologic phenomenon associated with ventricular arrhythmias, is usually detected from selected ECG leads. TWA amplitude measured in the 12-standard and the 3-orthogonal (vectorcardiographic) leads were compared here to identify which lead system yields a more adequate detection of TWA as a noninvasive marker for cardiac vulnerability to ventricular arrhythmias. Our adaptive match filter (AMF) was applied to exercise ECG tracings from 58 patients with an implanted cardiac defibrillator, 29 of which had ventricular tachycardia or fibrillation during follow-up (cases), while the remaining 29 were used as controls. Two kinds of TWA indexes were considered, the single-lead indexes, defined as the mean TWA amplitude over each lead (MTWAA), and lead-system indexes, defined as the mean and the maximum MTWAA values over the standard leads and over the orthogonal leads. Significantly (P < 0.05) higher TWA in the cases versus controls was identified only occasionally by the single-lead indexes (odds ratio: 1.0-9.9, sensitivity: 24-76%, specificity: 76-86%), and consistently by the lead-system indexes (odds ratio: 4.5-8.3, sensitivity: 57-72%, specificity: 76%). The latter indexes also showed a significant correlation (0.65-0.83) between standard and orthogonal leads. Hence, when using the AMF, TWA should be detected in all leads of a system to compute the lead-system indexes, which provide a more reliable TWA identification than single-lead indexes, and a better discrimination of patients at increased risk of cardiac instability. The standard and the orthogonal leads can be considered equivalent for TWA identification, so that TWA analysis can be limited to one-lead system.
Collapse
Affiliation(s)
- Laura Burattini
- Department of Information Engineering, Polytechnic University of Marche, Ancona, Italy.
| | | | | | | |
Collapse
|
29
|
Postexercise recovery of the spatial QRS/T angle as a predictor of sudden cardiac death. Heart Rhythm 2012; 9:1083-9. [DOI: 10.1016/j.hrthm.2012.02.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Indexed: 11/21/2022]
|
30
|
Raitoharju E, Seppälä I, Levula M, Kuukasjärvi P, Laurikka J, Nikus K, Huovila APJ, Oksala N, Klopp N, Illig T, Laaksonen R, Karhunen PJ, Viik J, Lehtinen R, Pelto-Huikko M, Tarkka M, Kähönen M, Lehtimäki T. Common variation in the ADAM8 gene affects serum sADAM8 concentrations and the risk of myocardial infarction in two independent cohorts. Atherosclerosis 2011; 218:127-33. [DOI: 10.1016/j.atherosclerosis.2011.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/05/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
|
31
|
Kenttä T, Karsikas M, Junttila MJ, Perkiömäki JS, Seppänen T, Kiviniemi A, Nieminen T, Lehtimäki T, Nikus K, Lehtinen R, Viik J, Kähönen M, Huikuri HV. QRS-T morphology measured from exercise electrocardiogram as a predictor of cardiac mortality. Europace 2010; 13:701-7. [PMID: 21186225 DOI: 10.1093/europace/euq461] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Total cosine R-to-T (TCRT) measured from the standard 12-lead electrocardiogram (ECG) reflects the spatial relationship between depolarization and repolarization wavefronts and a low TCRT value is a marker of poor prognosis. We tested the hypothesis that measurement of TCRT or QRS/T angle from exercise ECG would provide even more powerful prognostic information. METHODS AND RESULTS The prognostic significances of TCRT and QRS/T angle were assessed from exercise ECG recordings in 1297 patients [age 56 ± 13 years (mean ± SD), 67% males] undergoing a clinically indicated bicycle stress-test and the subsequent follow-up. During an average follow-up of 45 ± 12 months, 74 patients died (5.7%); 34 (2.6%) were cardiac deaths, and 24 (1.9%) were sudden cardiac deaths. Total cosine R-to-T and QRS/T angle exhibited a correlation with the RR intervals in the total cohort, but the individual responses were variable, e.g. median correlation of TCRT-RR was 0.89 with an inter-quartile range from 0.55 to 0.98. A reduced correlation of TCRT-RR during the recovery phase of exercise ECG predicted cardiac death [adjusted heart rate (HR) 3.5, 95% confidence interval (CI): 1.8-6.8, P= 0.001] similarly as the baseline TCRT measured from ECG at rest (adjusted HR 3.4, 95% CI: 1.4-8.1, P= 0.01). The poor correlation between the TCRT-RR both during the exercise and recovery was specifically related to a risk of sudden cardiac death (adjusted HR 6.2, 95% CI: 2.1-17.8, P< 0.001). CONCLUSIONS Loss of rate-adaptation of the spatial relationship between depolarization and repolarization wavefronts is a strong predictor of cardiac death, especially of sudden cardiac death.
Collapse
Affiliation(s)
- Tuomas Kenttä
- Division of Cardiology, Institute of Clinical Medicine, University of Oulu, Oulu, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Leino J, Verrier RL, Minkkinen M, Lehtimäki T, Viik J, Lehtinen R, Nikus K, Kööbi T, Turjanmaa V, Kähönen M, Nieminen T. Importance of regional specificity of T-wave alternans in assessing risk for cardiovascular mortality and sudden cardiac death during routine exercise testing. Heart Rhythm 2010; 8:385-90. [PMID: 21056698 DOI: 10.1016/j.hrthm.2010.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/01/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND T-wave alternans (TWA) indicates increased risk for life-threatening arrhythmias. However, the regional distribution and predictivity of TWA among precordial leads remain unknown. OBJECTIVE We analyzed the magnitude and prognostic power of TWA in precordial leads separately and in combination during routine exercise stress testing in the largest TWA study conducted to date. METHODS The Finnish Cardiovascular Study (FINCAVAS) enrolled consecutive patients (n = 3,598, 56 ± 13 [mean ± standard deviation] years old, 2,164 men, 1,434 women) with a clinically indicated exercise test with bicycle ergometer. TWA was analyzed with the time-domain modified moving average method. RESULTS During a follow-up of 55 months (interquartile range of 35-78 months), 231 patients died; 97 deaths were cardiovascular, and 46 were classified as sudden cardiac deaths (SCDs). In Cox analysis after adjustment for common coronary risk factors, each 20-μV increase in TWA in leads V1-V6 multiplied the hazard ratio for cardiovascular mortality by 1.486-fold (95% confidence interval [CI] 1.127-1.952; P = .005). Each 20-μV increase in TWA in lead V5 amplified the hazard ratio for cardiovascular mortality by 1.545 (95% CI 1.150-2.108; P = .004) and for SCD by 1.576 (95% CI 1.041-2.412; P = .033). CONCLUSIONS Maximum TWA monitored from anterolateral precordial lead V5 is the strongest predictor of cardiovascular mortality and SCD during routine exercise testing in our analysis. Higher TWA values indicate greater cardiovascular mortality and SCD risk, supporting the concept that quantification of TWA should receive more attention.
Collapse
|
33
|
Hernesniemi JA, Anttila K, Nieminen T, Kähönen M, Mononen N, Nikus K, Turjanmaa V, Viik J, Lehtinen R, Lehtimäki T. IL-18 gene polymorphism, cardiovascular mortality and coronary artery disease. Eur J Clin Invest 2010; 40:994-1001. [PMID: 20735470 DOI: 10.1111/j.1365-2362.2010.02356.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Interleukin 18(IL-18) is a pro-atherosclerotic cytokine. Elevated IL-18 levels and the genetic variation of the IL-18 have been previously linked with acute coronary events and cardiovascular mortality among patients with coronary artery disease (CAD). We studied the possible association between the IL-18 gene polymorphism and cardiovascular mortality during follow-up among Finnish patients who had undergone a clinical exercise stress test, in addition to the possible effect on the expression of angiography-verified CAD. MATERIALS AND METHODS A total of 2152 patients of the Finnish Cardiovascular Study (cohort study) were followed up for 6·3years and cardiovascular mortality was recorded. Angiography was performed on 461 patients. Genotyping of five common single nucleotide polymorphisms (SNPs) of the IL-18 gene was performed using the 5'nuclease assay for allelic discrimination with the ABI Prism 7900HT Sequence Detection System. RESULTS Among the study population, IL-18 gene polymorphism did not associate with cardiovascular mortality. According to adjusted binary regression analysis, the male carriers of one major haplotype (the only ones carrying the t allele of the +127 C/t SNP) had a lower occurrence rate for significant CAD defined as > 50% stenosis in at least one of the main branches of the coronary arteries (OR 0·495, 95% CI 0·862-0·284, P=0·041). No associations were observed among women. The sex-by-genotype interaction was significant (P=0·033). CONCLUSIONS The IL-18 gene was not found to associate significantly with mortality. Among patients who had coronary angiography, one major haplotype of the IL-18 gene has a gender-dependent different impact on the expression of CAD.
Collapse
Affiliation(s)
- Jussi A Hernesniemi
- Laboratory of Atherosclerosis Genetics, Department of Clinical Chemistry, Tampere University Hospital, Centre for Laboratory Medicine, Tampere, Finland Medical School, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Atrioventricular conduction and cardiovascular mortality: Assessment of recovery PR interval is superior to pre-exercise measurement. Heart Rhythm 2010; 7:796-801. [DOI: 10.1016/j.hrthm.2010.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 02/20/2010] [Indexed: 11/23/2022]
|
35
|
Puurtinen M, Nieminen T, Kähönen M, Lehtimäki T, Lehtinen R, Nikus K, Hyttinen J, Viik J. Value of leads V4R and CM5 in the detection of coronary artery disease during exercise electrocardiographic test. Clin Physiol Funct Imaging 2010; 30:308-12. [PMID: 20497446 DOI: 10.1111/j.1475-097x.2010.00942.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The usefulness of the right precordial unipolar leads and the value of the bipolar lead CM5 in the detection of coronary artery disease (CAD) with exercise electrocardiographic (ECG) test are not well documented. The objective of this study was to evaluate the diagnostic performance of leads V4R and CM5. The study population comprised 579 patients referred for a bicycle exercise ECG test in the Finnish Cardiovascular Study. Patients were divided into three groups: angiographically proven CAD (CAD, n = 255), no CAD by angiography (NoCAD, n = 126), and low likelihood of CAD (LLC, n = 198). The maximum ST-segment depression at peak exercise was used as a parameter, and the diagnostic accuracy of different leads was assessed by receiver operating characteristic (ROC) analysis. Sensitivity and specificity values at a cut-off criterion of -0.10 mV ST-segment, 1-mm ST depression, were determined. According to the results, incorporating lead V4R with the standard leads decreased the ROC area from 0.71 to 0.69 (comparison CAD versus LLC) and from 0.55 to 0.53 (comparison CAD versus NoCAD) and had no effect on sensitivity or specificity. Adding lead CM5 to the standard leads did not affect the ROC area but increased the sensitivity and decreased the specificity. In conclusion, the use of right precordial lead V4R along with the standard 12-lead system does not improve the performance of the exercise ECG in diagnosing CAD. Adding lead CM5 to the standard leads increases the sensitivity but does not change the overall diagnostic performance.
Collapse
Affiliation(s)
- Merja Puurtinen
- Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Svart K, Lehtinen R, Nieminen T, Nikus K, Lehtimäki T, Kööbi T, Niemelä K, Niemi M, Turjanmaa V, Kähönen M, Viik J. Exercise electrocardiography detection of coronary artery disease by ST-segment depression/heart rate hysteresis in women: The Finnish Cardiovascular Study. Int J Cardiol 2010; 140:182-8. [DOI: 10.1016/j.ijcard.2008.11.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 08/08/2008] [Accepted: 11/08/2008] [Indexed: 11/15/2022]
|
37
|
Combined assessment of heart rate recovery and T-wave alternans during routine exercise testing improves prediction of total and cardiovascular mortality: The Finnish Cardiovascular Study. Heart Rhythm 2009; 6:1765-71. [DOI: 10.1016/j.hrthm.2009.08.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2009] [Accepted: 08/12/2009] [Indexed: 11/21/2022]
|
38
|
MINKKINEN MIKKO, KÄHÖNEN MIKA, VIIK JARI, NIKUS KJELL, LEHTIMÄKI TERHO, LEHTINEN RAMI, KÖÖBI TIIT, TURJANMAA VÄINÖ, KAISER WILLI, VERRIER RICHARDL, NIEMINEN TUOMO. Enhanced Predictive Power of Quantitative TWA during Routine Exercise Testing in the Finnish Cardiovascular Study. J Cardiovasc Electrophysiol 2009; 20:408-15. [DOI: 10.1111/j.1540-8167.2008.01325.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Leino J, Virtanen M, Kähönen M, Nikus K, Lehtimäki T, Kööbi T, Lehtinen R, Turjanmaa V, Viik J, Nieminen T. Exercise-test-related heart rate variability and mortality: the Finnish Cardiovascular Study. Int J Cardiol 2009; 144:154-5. [PMID: 19167108 DOI: 10.1016/j.ijcard.2008.12.123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 12/14/2008] [Indexed: 11/25/2022]
|
40
|
Minkkinen M, Nieminen T, Verrier RL, Leino J, Lehtimäki T, Viik J, Lehtinen R, Nikus K, Kööbi T, Turjanmaa V, Kähönen M. Impaired exercise capacity predicts sudden cardiac death in a low-risk population: enhanced specificity with heightened T-wave alternans. Ann Med 2009; 41:380-9. [PMID: 19301163 DOI: 10.1080/07853890902802971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AIMS Because sudden cardiac death (SCD) is due to cardiac electrical instability, we postulated that prediction of this mode of death by exercise capacity will be enhanced by combined assessment with T-wave alternans (TWA), an index of repolarization abnormality. MATERIAL AND METHODS The Finnish Cardiovascular Study enrolled consecutive patients (n=2,044) with a routine clinically indicated exercise test. Exercise capacity was measured in metabolic equivalents (METs) and TWA by time-domain modified moving average method. RESULTS During 47.2+/-12.8-month follow-up (mean+/-SD) 120 patients died; 58 were cardiovascular deaths, and 29 were SCD. In multivariate analysis after adjustment for sex, age, smoking, use of beta-blockers, as well as other common coronary risk factors, the relative risk of patients whose exercise capacity was depressed (MET < 8) was 8.8 (95% CI 2.0-38.9, P=0.004) for SCD. The combination of low exercise capacity (MET < 8) and elevated TWA (> or =65 microV) yielded relative risks for SCD of 36.1 (6.3-206.0, P<0.001), for cardiovascular mortality of 21.1 (6.7-66.2, P<0.001), and for all-cause mortality of 7.8 (3.5-17.4, P<0.001) over patients with neither factor. CONCLUSIONS Reduced exercise capacity, particularly in combination with heightened TWA, indicating enhanced cardiac electrical instability, powerfully predicts risk for SCD in patients referred for exercise testing.
Collapse
|
41
|
Koskela J, Kähönen M, Fan M, Nieminen T, Lehtinen R, Viik J, Nikus K, Niemelä K, Kööbi T, Turjanmaa V, Pörsti I, Lehtimäki T. Effect of common KCNE1 and SCN5A ion channel gene variants on T-wave alternans, a marker of cardiac repolarization, during clinical exercise stress test: the Finnish Cardiovascular Study. Transl Res 2008; 152:49-58. [PMID: 18674739 DOI: 10.1016/j.trsl.2008.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 06/18/2008] [Accepted: 06/20/2008] [Indexed: 12/19/2022]
Abstract
T-wave alternans (TWA) in electrocardiography (ECG) is a marker of cardiac repolarization, the molecular regulation of which is incompletely understood. High TWA and prolonged QT intervals are both associated with ventricular arrhythmias and sudden death. Therefore, we tested the hypothesis of whether the same mutations that influence the QT interval also affect TWA variation. We examined the effect of 3 ion channel gene single nucleotide polymorphisms (SNPs), rs1805127, rs727957 KCNE1, and rs1805124 SCN5A, on TWA during a clinical exercise test. A total of 2008 subjects from the Finnish Cardiovascular Study underwent an exercise test with online ECG recording. TWA was measured by using the time-domain, modified moving average method. Maximum values at rest, during maximal exercise, and during recovery were used as outcome measures in statistical analysis. Moreover, 4-year survival data were collected and ion channel SNPs were determined. TWA was lowest in subjects with the TT genotype of rs1805127 during all phases of the exercise test (RANOVA main effect for genotype, P = 0.018). The result remained significant after adjustment for age, existing coronary heart disease, and beta-blocker medication status (RANCOVA, P = 0.035). Of the polymorphisms studied, only rs1805127 had a significant association with mortality (P = 0.047). The most common G-C haplotype, formed by rs727957 and rs1805127, was associated with TWA (RANOVA, P = 0.007) but not with mortality. The rs1805124 polymorphism was not associated with TWA. The common KCNE1 gene variant rs1805127 is associated with TWA during an exercise test in a Finnish population, which provides additional evidence that KCNE1 genetics may influence cardiac repolarization and cardiovascular mortality.
Collapse
Affiliation(s)
- Jenni Koskela
- Department of Internal Medicine, Tampere University Hospital and Medical School at the University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tobin MD, Kähönen M, Braund P, Nieminen T, Hajat C, Tomaszewski M, Viik J, Lehtinen R, Ng GA, Macfarlane PW, Burton PR, Lehtimäki T, Samani NJ. Gender and effects of a common genetic variant in the NOS1 regulator NOS1AP on cardiac repolarization in 3761 individuals from two independent populations. Int J Epidemiol 2008; 37:1132-41. [PMID: 18511491 DOI: 10.1093/ije/dyn091] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND A longer heart-rate corrected QT interval (QTc) is associated with increased risk of ventricular arrhythmias. Women have longer resting QTc and are more likely than men to develop drug-induced QT prolongation. Recent studies have shown association between resting QTc and a common variant (rs10494366) of the NOS1 regulator, NOS1AP. We investigated the association between rs10494366 in NOS1AP and QTc, and assessed gender-specific NOS1AP associations with QTc during rest and after exercise. METHODS We investigated the SNP associations with resting QTc in 919 women and 918 men from 504 representative families in the UK GRAPHIC study, and with QTc at rest and at 3 min recovery after exercise in 699 women and 1225 men referred for exercise testing in the Finnish FINCAVAS study. RESULTS In the GRAPHIC study the minor allele (G) of the NOS1AP SNP rs10494366 prolonged QTc by 4.59 ms (95% CI 2.77-6.40; P = 7.63/10(7)) in women, but only by 1.62 ms (95% CI -0.15 to 3.38; P = 0.073) in men (gender-SNP interaction term P = 0.025). In the FINCAVAS study the G allele significantly prolonged QTc in both women (P = 0.0063) and men (P = 0.0043) at 3 min recovery after exercise, but at rest an association was only seen in women (P = 0.020 excluding outliers). CONCLUSIONS A common NOS1AP variant prolongs QTc with a difference between genders. Further studies should aim to confirm this finding and to assess whether NOS1AP genotype influences the risk of drug-induced QT prolongation and risk of consequent arrhythmias.
Collapse
Affiliation(s)
- Martin D Tobin
- Department of Health Sciences, University of Leicester, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Nieminen T, Leino J, Maanoja J, Nikus K, Viik J, Lehtimäki T, Kööbi T, Lehtinen R, Niemelä K, Turjanmaa V, Kähönen M. The prognostic value of haemodynamic parameters in the recovery phase of an exercise test. The Finnish Cardiovascular Study. J Hum Hypertens 2008; 22:537-43. [PMID: 18509348 DOI: 10.1038/jhh.2008.38] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We tested the hypothesis that the change from the peak to recovery values of systolic arterial pressure (SAP recovery) and rate-pressure product (RPP recovery) can be used to predict all-cause and cardiovascular mortality, as well as sudden cardiac death (SCD) in patients referred to a clinical exercise stress test. As a part of the Finnish Cardiovascular Study (FINCAVAS), consecutive patients (n=2029; mean age+/-SD=57+/-13 years; 1290 men and 739 women) with a clinically indicated exercise test using a bicycle ergometer were included in the present study. Capacities of attenuated SAP recovery, RPP recovery and heart rate recovery (HRR) to stratify the risk of death were estimated. During a follow-up (mean+/-s.d.) of 47+/-13 months, 122 patients died; 58 of the deaths were cardiovascular and 33 were SCD. In Cox regression analysis after adjustment for the peak level of the variable under assessment, age, sex, use of beta-blockers, previous myocardial infarction and other common coronary risk factors, the hazard ratio of the continuous variable RPP recovery (in units 1000 mm Hg x b.p.m.) was 0.85 (95% CI: 0.73-0.98) for SCD, 0.87 (0.78-0.97) for cardiovascular mortality, and 0.87 (0.81 to 0.94) for all-cause mortality. SAP recovery was not a predictor of mortality. The relative risks of having HRR below 18 b.p.m., a widely used cutoff point, were as follows: for SCD 1.28 (0.59-2.81, ns), for cardiovascular mortality 2.39 (1.34-4.26) and for all-cause mortality 2.40 (1.61-3.58). In conclusion, as a readily available parameter, RPP recovery is a promising candidate for a prognostic marker.
Collapse
Affiliation(s)
- T Nieminen
- Department of Pharmacological Sciences, Medical School, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Koskela J, Laiho J, KäHönen M, Rontu R, Lehtinen R, Viik J, Niemi M, Niemelä K, Kööbi T, Turjanmaa V, Pörsti I, Lehtimäki T, Nieminen T. Potassium channel KCNH2 K897T polymorphism and cardiac repolarization during exercise test: The Finnish Cardiovascular Study. Scandinavian Journal of Clinical and Laboratory Investigation 2008; 68:31-8. [PMID: 17852802 DOI: 10.1080/00365510701496488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Cardiac repolarization is regulated, in part, by the KCNH2 gene, which encodes a rapidly activating component of the delayed rectifier potassium channel. The gene expresses a functional single nucleotide polymorphism, K897T, which changes the biophysical properties of the channel. The objective of this study was to evaluate whether this polymorphism influences two indices of repolarization--the QT interval and T-wave alternans (TWA)--during different phases of a physical exercise test. MATERIAL AND METHODS The cohort consisted of 1,975 patients undergoing an exercise test during which on-line electrocardiographic data were registered. Information on coronary risk factors and medication was recorded. The 2690A>C nucleotide variation in the KCNH2 gene corresponding to the K897T amino acid change was analysed after polymerase chain reaction with allele-specific TaqMan probes. RESULTS Among all subjects, the QTc intervals did not differ between the three genotype groups (p> or =0.31, RANOVA). Women with the CC genotype tended to have longer QT intervals during the exercise test, but the difference was statistically significant only at rest (p = 0.011, ANOVA). This difference was also detected when the analysis was adjusted for several factors influencing the QT interval. No statistically significant effects of the K897T polymorphism on TWA were observed among all subjects (p = 0.16, RANOVA), nor in men and women separately. CONCLUSIONS The K897T polymorphism of the KCNH2 gene may not be a major genetic determinant for the TWA, but the influence of the CC genotype on QT interval deserves further research among women.
Collapse
Affiliation(s)
- J Koskela
- Internal Medicine, Tampere University Hospital and Tampere University Medical School, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lorgis L, Zeller M, Beer JC, Lagrost AC, Buffet P, L'Huillier I, Sicard P, Cottin Y. [Epidemiology of acute coronary syndrome in Europe]. Ann Cardiol Angeiol (Paris) 2007; 56 Suppl 1:S2-7. [PMID: 17719353 DOI: 10.1016/s0003-3928(07)80020-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Epidemiological data concerning acute coronary syndromes in Europe are based on national registries, studies by the European Society of Cardiology within the framework of the EuroHeart Survey and on the study of European population sub-groups in large international cohorts. In this article, recently published studies will be reviewed, and the principal developments in different countries as well as the characteristics and particularities of the most recent epidemiological data will be highlighted. In Europe, the presentation of acute coronary syndromes (ACS) has evolved considerably over the last ten years. This evolution is characterized by a reduction in the proportion of acute coronary syndromes with ST-segment elevation (STEMI) and by ageing populations.
Collapse
Affiliation(s)
- L Lorgis
- Service de cardiologie, CHU Bocage, boulevard Mal de Lattre de Tassigny, 21034 Dijon, France
| | | | | | | | | | | | | | | |
Collapse
|