1
|
Drobnik S, Scharnagl H, Samani NJ, Braund PS, Nelson CP, Hollstein T, Kassner U, Dressel A, Drobnik W, März W. Evaluation of current indirect methods for measuring LDL-cholesterol. Clin Chem Lab Med 2025; 63:1099-1108. [PMID: 39964360 DOI: 10.1515/cclm-2025-0024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/29/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVES Accurately quantifying low-density lipoprotein cholesterol (LDL-C) is crucial for precise cardiovascular disease risk assessment and treatment decisions. The commonly used Friedewald equation (LDL-CFW) has faced criticism for its tendency to underestimate LDL-C, particularly at high triglycerides (TG) or low LDL-C, potentially leading to undertreatment. Newer equations, such as those by Martin and Hopkins (LDL-CMH) or Sampson (LDL-CSN), have been proposed as alternatives. Our study aimed to assess the validity of LDL-CFW, LDL-CMH, and LDL-CSN compared to ß-quantification (LDL-CUC), the reference method recommended by the Lipid Research Clinics. METHODS Using data from three studies comprising 5,738 datasets, LDL-C was determined with the four methods in samples with TG up to 5.65 mmol/L. We calculated median and mean differences, correlations, and used the Passing and Bablok regression for comparisons. Concordance/discordance analyses were conducted. RESULTS All equations provided generally accurate LDL-C estimations with slight differences among them. At TG<1.69 mmol/L, no clinically significant divergences were observed. As TG values increased, LDL-CFW offered the most accurate estimation, followed by LDL-CSN, while LDL-CMH exhibited increasingly strong positive bias. LDL-CFW was not inferior to LDL-CSN and LDL-CMH in terms of concordance/discordance. CONCLUSIONS LDL-CFW generally provided reliable estimates of LDL-C in most samples, showing non-inferiority to LDL-CSN or LDL-CMH, thereby confirming its legitimacy for routine use. Since current treatment recommendations are based on studies employing LDL-CFW, its replacement by alternatives is not justified.
Collapse
Affiliation(s)
- Sophia Drobnik
- Medical Clinic I, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Tim Hollstein
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ursula Kassner
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Dressel
- D•A•CH-Gesellschaft Prävention von Herz-Kreislauf-Erkrankungen e.V., Hamburg, Germany
- Dr.Dressel Consulting, Mannheim, Germany
| | | | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
- Department of Internal Medicine III (Cardiology, Angiology, Pneumology), Medical Faculty Heidelberg, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
2
|
Daschner C, Kleber ME, Ayasse N, Stach K, Yücel G, Husain-Syed F, Niessner A, Krüger B, März W, Krämer BK, Yazdani B. Central vs. Brachial Blood Pressure and Pulse Pressure Amplification for Mortality Risk Prediction in Patients Undergoing Coronary Angiography. Am J Hypertens 2025; 38:272-279. [PMID: 39699029 DOI: 10.1093/ajh/hpae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/20/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Arterial hypertension is a significant risk factor for cardiovascular (CV) morbidity and mortality. Although central blood pressure (BP) evaluation is considered the gold standard, the reliability of non-invasive measurements remains unclear. Therefore, we compared the predictive value of invasively measured central BP with non-invasively measured brachial BP and analyzed pulse pressure (PP) amplification (delta-PP; the difference between central and peripheral PP) as an independent predictor of mortality. METHODS We analyzed systolic BP (SBP), diastolic BP (DBP), mean arterial BP (MAP), PP, and delta-PP as predictors of CV and all-cause mortality in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, involving 3,316 patients referred for coronary angiography. RESULTS All brachial BP parameters, except DBP, were significantly linked to all-cause and CV mortality in a univariate analysis. A 10 mm Hg increase in SBP, MAP, and PP corresponded to increased risks of all-cause (11%, 10%, and 19%) and CV mortality (11%, 11%, and 18%). Central SBP and PP showed similar, but numerically weaker, associations with increased risks of all-cause (5% and 10%) and CV mortality (4% and 8%). After adjusting for age, sex, body mass index, diabetes mellitus, and eGFR, only delta-PP independently predicted mortality with a 10 mm Hg increase linked to a 4% reduction in all causes and a 6% reduction in CV mortality. CONCLUSIONS Neither brachial nor centrally measured BP parameters were independent mortality predictors in contrast to PP amplification, which remained an independent predictor of mortality in multivariate analysis, in a cohort with a medium to high CV risk profile. As PP amplification decreased, mortality increased.
Collapse
Affiliation(s)
- Clara Daschner
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Baden-Württemberg and Bayern, Germany
| | - Niklas Ayasse
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
| | - Ksenija Stach
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
| | - Gökhan Yücel
- First Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
| | - Faeq Husain-Syed
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Giessen, Hessen, Germany
| | - Alexander Niessner
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Second Medical Department with Cardiology and Intensive Care Medicine, Klinik Landstrasse, Vienna, Vienna, Austria
| | - Bernd Krüger
- Third department of Medicine, Hospital Darmstadt, Darmstadt, Hessen, Germany
| | - Winfried März
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Baden-Württemberg and Bayern, Germany
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Steiermark, Austria
| | - Bernhard K Krämer
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
- Center for Preventive Medicine Baden-Württemberg (CPMBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Baden-Württemberg, Germany
- European Center for Angioscience ECAS, Faculty of Medicine of the University of Heidelberg, Mannheim, Baden-Württembarg, Germany
| | - Babak Yazdani
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Baden-Württemberg, Germany
| |
Collapse
|
3
|
Weyrich M, Zewinger S, Sarakpi T, Rasper T, Kleber ME, Cremer S, Zanders L, Fleck F, Siegbahn A, Wallentin L, Abplanalp WT, Nerbas L, Fay S, Eberle AL, Dimmeler S, März W, Speer T, Zeiher AM. Mosaic loss of Y chromosome and mortality after coronary angiography. Eur Heart J 2025:ehaf035. [PMID: 39935193 DOI: 10.1093/eurheartj/ehaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/30/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND AND AIMS Acquired somatic mutations emerged as important drivers of adverse cardiovascular disease outcomes. Recently, mosaic loss of Y chromosome (LOY) in haematopoietic cells was identified to induce diffuse cardiac fibrosis in male mice. The aim of the present study was to determine the association between LOY and cardiovascular mortality in patients undergoing coronary angiography. METHODS LOY was quantified in 1698 male participants of the LURIC study, who underwent coronary angiography, and its association with all-cause and cardiovascular mortality was determined. Furthermore, the interaction between LOY and inherited genetic susceptibility for cardiac fibrosis was assessed. RESULTS The frequency of LOY steeply increased in male participants of LURIC at the age of 60 years. Loss of Y chromosome > 17% was associated with significantly higher all-cause [hazard ratio (HR) 1.41, 95% confidence interval (CI) 1.09-1.82] and cardiovascular mortality (HR 1.49, 95% CI 1.09-2.03), which was driven by a higher risk for fatal myocardial infarction (HR 2.65, 95% CI 1.46-4.81). Loss of Y chromosome > 17% was associated with a profibrotic and proinflammatory plasma protein expression profile as characterized by higher plasma levels of osteoprotegerin, matrix metalloproteinase-12, growth differentiation factor 15, heparin-binding EGF-like growth factor, and resistin. Genetic predisposition for lower myocardial fibrosis attenuated the association between LOY and cardiovascular mortality. Genome-wide methylation analyses identified differential methylation in 298 genes including ACTB, RPS5, WDR1, CD151, and ARAP1. Single-cell RNA sequencing further confirmed differential gene expression of 37 of these genes in LOY in peripheral blood mononuclear cells comprising a set of fibrosis-regulating genes including RPS5. RPS5 silencing in macrophages induced a paracrine induction of collagen expression in cardiac fibroblasts documenting a functional role in vitro. CONCLUSIONS LOY represents an important independent risk factor for cardiovascular mortality in male patients with coronary artery disease. Targeting LOY may represent a sex-specific personalized medicine approach.
Collapse
Affiliation(s)
- Michael Weyrich
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Stephen Zewinger
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Hôpital Robert Schumann, Hôpital Kirchberg, Luxemburg, Luxemburg
- Medical Faculty, Saarland University, Homburg/Saar, Germany
| | - Tamim Sarakpi
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Tina Rasper
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Marcus E Kleber
- Vth Department of Medicine, University of Heidelberg, University Medical Center, Medical Faculty Mannheim, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Sebastian Cremer
- Department of Medicine, Cardiology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Lukas Zanders
- Department of Medicine, Cardiology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Fenja Fleck
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Wesley Tyler Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Linda Nerbas
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Sandra Fay
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Aaron L Eberle
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Winfried März
- Vth Department of Medicine, University of Heidelberg, University Medical Center, Medical Faculty Mannheim, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Holding Deutschland GmbH, SYNLAB Academy, Mannheim, Germany
| | - Thimoteus Speer
- Department of Internal Medicine 4, Nephrology, Goethe University Frankfurt, University Hospital, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- Else Kroener-Fresenius Center for Nephrological Research, Goethe University Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| | - Andreas M Zeiher
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research DZHK, Partner Site Frankfurt, Theodor-Stern-Kai 7, Frankfurt 60590, Germany
| |
Collapse
|
4
|
Herrmann M, Keppel MH, Zelzer S, Alonso N, Cavalier E, Kleber M, Enko D, Scharnagl H, Pilz S, März W. The role of functional vitamin D deficiency and low vitamin D reservoirs in relation to cardiovascular health and mortality. Clin Chem Lab Med 2025; 63:208-219. [PMID: 38890759 DOI: 10.1515/cclm-2024-0391] [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: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVES The role of vitamin D deficiency in cardiovascular disease (CVD) is controversial. Inherent biological and analytical limitations compromise the specificity of widely used 25-hydroxyvitamin D [25(OH)D] cut-offs. Simultaneous determination of 25(OH)D and 24,25-dihydroxyvitamin D [24,25(OH)2D] permits a functional assessment of vitamin D metabolism. The present study compared the associations of functional vitamin D deficiency and low vitamin D reservoirs with CVD mortality and CVD burden. METHODS 25(OH)D, 24,25(OH)2D, the degree of coronary obstruction on angiography, high-sensitive cardiac troponin T (hs-cTnT), N-terminal brain natriuretic peptide (NT-proBNP), and 10-year CVD mortality were obtained from 2,456 participants of the LURIC (Ludwigshafen Risk and Cardiovascular Health) study. RESULTS Neither low 25(OH)D concentrations nor functional vitamin D deficiency were associated with the number of atherosclerotic coronary arteries or the degree of coronary obstruction. Over a median follow-up of 9.9 years, 454 participants died (23.6 %) due to CVD. CVD mortality was doubled in individuals with 25(OH)D concentrations below the widely used cut-off for deficiency of <50 nmol/L [20 ng/mL] (21.6 vs. 11.5 %). In individuals with and without functional vitamin D deficiency, CVD mortality was 25.0 and 16.7 %, respectively. NT-proBNP and heart failure prevalence were also higher in vitamin D deficient individuals. CONCLUSIONS Vitamin D deficient individuals have markedly higher CVD mortality, but only marginally higher hs-cTnT concentrations. A higher prevalence of heart failure and higher NT-proBNP concentrations suggest a link between vitamin D deficiency and cardiac function. The traditional and metabolic assessment of vitamin D status showed comparable associations for the different parameters of cardiac health.
Collapse
Affiliation(s)
- Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Martin H Keppel
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Nerea Alonso
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liege, Liege, Belgium
| | - Marcus Kleber
- Department of Internal Medicine 5 (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Synlab Human Genetics Laboratory, Synlab AG, Mannheim, Germany
| | - Dietmar Enko
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Stefan Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Human Genetics Laboratory, Synlab AG, Mannheim, Germany
- Synlab Academy, Synlab Holding Germany GmbH, Mannheim, Germany
| |
Collapse
|
5
|
Daschner C, Kleber ME, Stach K, Yuecel G, Husain-Syed F, Ayasse N, Berg AH, März W, Krämer BK, Yazdani B. Association between Arterial Stiffness, Carbamylation, and Mortality in Patients Undergoing Coronary Angiography with No or Mild Chronic Kidney Disease. Cardiorenal Med 2024; 15:83-97. [PMID: 39701045 PMCID: PMC11844701 DOI: 10.1159/000543143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
INTRODUCTION The processes of atherosclerosis, inflammation, and carbamylation are closely linked in cardiovascular (CV) disease, but the potential of carbamylation burden as a CV mortality predictor is unclear, especially in patients with no or mild chronic kidney disease (CKD). This study aimed to investigate whether elevated carbamylated albumin (C-Alb), as a surrogate marker for carbamylation burden, is associated with mortality and arterial stiffness/atherosclerotic burden in patients with no or mild CKD, using pulse pressure (PP) as a marker for arterial stiffness. METHODS We measured C-Alb in 3,193 participants of the Ludwigshafen Risk and Cardiovascular Health study who had been referred for coronary angiography and followed up for 10 years. RESULTS The mean age was 62.7 years, and 30.4% were female. Mean blood pressure was 141/81 mm Hg, and mean C-Alb was 5.54 mmol/mol. Increase in C-Alb levels was associated with older age; female sex; increased PP, high-sensitivity C-reactive protein, and interleukin-6 levels; and increased incidence of coronary artery disease (CAD), peripheral artery disease (PAD), and carotid stenosis. In contrast, BMI, diastolic blood pressure (DBP), albumin, and the proportion of active smokers decreased with increasing C-Alb levels. In particular, C-Alb showed a highly significant correlation with CAD severity: Friesinger (Pearson correlation coefficient [r] = 0.082, p < 0.001) and Gensini score (r = 0.066, p < 0.001). The area under the curve (AUC) for all-cause mortality prediction by the European Society of Cardiology Heart Score (ESC-HS) significantly improved from 0.719 to 0.735, and the AUC for CV mortality prediction based on C-Alb increased from 0.726 to 0.750 in patients without previously known CV disease. C-Alb correlated directly and significantly with PP (r = 0.062, p < 0.001), which was consistently the strongest predictor of mortality across all C-Alb tertiles. The hazard ratios (HRs) for all-cause mortality per 10 mm Hg increase (or 1,000 mm Hg/min increase for double product [DP]) in the 1st tertile of C-Alb were 1.18, 1.13, 1.11, and 1.11 for PP, mean arterial pressure (MAP), systolic blood pressure (SBP), and DP, respectively, but the HR for DBP did not reach significance. In the 3rd tertile of C-Alb, the HRs were 1.13, 1.05, and 1.09, for PP, SBP, and DP, respectively, but the HR for MAP did not reach significance. CONCLUSION C-Alb may be a valuable biomarker for assessing CV risk and improving mortality prediction even in patients with no or mild CKD. The findings support the notion of a crosslink between carbamylation, inflammation, atherosclerosis, and mortality. While these results are promising, further research is needed to fully elucidate the role of C-Alb in CV disease progression and risk stratification.
Collapse
Affiliation(s)
- Clara Daschner
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Marcus E. Kleber
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Ksenija Stach
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Goekhan Yuecel
- First Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Faeq Husain-Syed
- Department of Medicine II, Division of Nephrology, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Niklas Ayasse
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Anders H. Berg
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Winfried März
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Augsburg, Germany
| | - Bernhard K. Krämer
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine Baden-Württemberg (CPMBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience ECAS, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Babak Yazdani
- Fifth Department of Medicine, University Medical Center Mannheim UMM, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine Baden-Württemberg (CPMBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
6
|
Zittermann A, Zelzer S, Herrmann M, Kleber M, Maerz W, Pilz S. Association between magnesium and vitamin D status in adults with high prevalence of vitamin D deficiency and insufficiency. Eur J Nutr 2024; 64:48. [PMID: 39680162 PMCID: PMC11649730 DOI: 10.1007/s00394-024-03559-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE It has been assumed that magnesium (Mg) status may interact with vitamin D status. We therefore aimed at investigating the association between Mg and vitamin D status in a large cohort of adult individuals with a high prevalence of deficient/insufficient vitamin D and Mg status. METHODS We used data from the Ludwigshafen Risk and Cardiovascular Health Study (n = 2,286) to analyze differences according to serum Mg status in circulating 25-hydroxyvitamin D [25(OH)D] (primary endpoint), 24,25-dihydroxyvitamin D3 [24,25(OH)2D3], vitamin D metabolite ratio and calcitriol, and odds ratios for deficient or insufficient 25(OH)D (secondary endpoints). We performed unadjusted and risk score (RS) adjusted and matched analyses. RESULTS Of the study cohort (average age > 60 years), one third was 25(OH)D deficient (< 12 ng/mL), one third 25(OH)D insufficient (12 to < 20 ng/mL), about 10% Mg deficient (< 0.75 mmol/L) and additional 40% potentially Mg deficient (0.75 to 0.85 mmol/L). In adjusted/matched analyses, 25(OH)D was only non-significantly lower in Mg deficient or insufficient groups versus their respective control group (P > 0.05). Only the RS-adjusted, but not the RS-matched odds ratio of 25(OH)D deficiency was significantly lower for the group with adequate versus deficient/potentially deficient Mg status (0.83; 95%CI: 0.69-0.99), and only the RS-matched, but not the RS-adjusted odds ratio of 25(OH)D insufficiency was significantly lower for non-deficient versus deficient Mg status (0.69; 95%CI: 0.48-0.99). Other adjusted or matched secondary endpoints did not differ significantly between subgroups of Mg status. CONCLUSIONS Our data indicate only little effect between Mg and vitamin D status in adults with high prevalence of vitamin D deficiency and insufficiency.
Collapse
Affiliation(s)
- Armin Zittermann
- Clinic for Thoracic and Cardiovascular Surgery, Herz-und Diabeteszentrum NRW, Georgstr. 11, D-32545, Bad Oeynhausen, Germany.
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, 8036, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, 8036, Austria
| | - Marcus Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology, Lipidology), Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, 68163, Mannheim, Germany
| | - Winfried Maerz
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology, Lipidology), Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, 68163, Mannheim, Germany
- SYNLAB Holding, Deutschland GmbH, 68159, Mannheim, Augsburg, Germany
| | - Sefan Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, 8036, Austria
| |
Collapse
|
7
|
Ciantar J, Marttila S, Rajić S, Kostiniuk D, Mishra PP, Lyytikäinen LP, Mononen N, Kleber ME, März W, Kähönen M, Raitakari O, Lehtimäki T, Raitoharju E. Identification and functional characterisation of DNA methylation differences between East- and West-originating Finns. Epigenetics 2024; 19:2397297. [PMID: 39217505 PMCID: PMC11382697 DOI: 10.1080/15592294.2024.2397297] [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: 04/29/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Eastern and Western Finns show a striking difference in coronary heart disease-related mortality; genetics is a known contributor for this discrepancy. Here, we discuss the potential role of DNA methylation in mediating the discrepancy in cardiometabolic disease-risk phenotypes between the sub-populations. We used data from the Young Finns Study (n = 969) to compare the genome-wide DNA methylation levels of East- and West-originating Finns. We identified 21 differentially methylated loci (FDR < 0.05; Δβ >2.5%) and 7 regions (smoothed FDR < 0.05; CpGs ≥ 5). Methylation at all loci and regions associates with genetic variants (p < 5 × 10-8). Independently of genetics, methylation at 11 loci and 4 regions associates with transcript expression, including genes encoding zinc finger proteins. Similarly, methylation at 5 loci and 4 regions associates with cardiometabolic disease-risk phenotypes including triglycerides, glucose, cholesterol, as well as insulin treatment. This analysis was also performed in LURIC (n = 2371), a German cardiovascular patient cohort, and results replicated for the association of methylation at cg26740318 and DMR_11p15 with diabetes-related phenotypes and methylation at DMR_22q13 with triglyceride levels. Our results indicate that DNA methylation differences between East and West Finns may have a functional role in mediating the cardiometabolic disease discrepancy between the sub-populations.
Collapse
Affiliation(s)
- Joanna Ciantar
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Saara Marttila
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Gerontology Research Center, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Sonja Rajić
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Daria Kostiniuk
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, 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
| | - Emma Raitoharju
- Molecular Epidemiology (MOLE), 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
- Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
8
|
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
|
9
|
Berger M, März W, Niessner A, Delgado G, Kleber M, Scharnagl H, Marx N, Schuett K. IL-6 and hsCRP predict cardiovascular mortality in patients with heart failure with preserved ejection fraction. ESC Heart Fail 2024; 11:3607-3615. [PMID: 39003598 PMCID: PMC11631318 DOI: 10.1002/ehf2.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 07/15/2024] Open
Abstract
AIMS Inflammation accompanies heart failure (HF) and elevated levels of inflammatory biomarkers are linked to new onset of HF. However, whether the prognostic relevance of inflammatory biomarkers is different in HF with reduced (HFrEF) and preserved ejection fraction (HFpEF) is unclear. The aim of the current study is to explore the role of inflammation on the mortality risk in patients with HF. METHODS We analysed interleukin-6 and hsCRP levels by ELISA and immunonephelometry, respectively, in HFpEF and HFrEF patients referred for coronary angiography and assessed the prognostic value in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. RESULTS HF was present in 1086 patients (N = 506 HFpEF; N = 580 HFrEF; mean age 65 ± 10 years; 28% female). Increasing IL-6 levels were significantly associated with increased CV mortality in HFpEF [1.5 (95% CI: 1.1-2.2), P = 0.018] but not HFrEF [HR 1.3 (95% CI: 1.0-1.7), P = 0.06] patients. High-sensitive CRP followed a similar pattern but failed to reach statistical significance after full-adjustment (HFpEF: HR 1.4 95%C I: 1.0-2.0; P = 0.065; HFrEF HR: 1.0 95% CI: 0.7-1.3; P = 0.800). Interaction analysis in patients stratified by IL-6 and N terminal pro brain natriuretic peptide (NT-proBNP) above and below the median revealed a stepwise increase in CV-mortality in HFpEF (P = 0.036) but not HFrEF patients (P = 0.220). To investigate the relationship between IL-6 and NT-proBNP, we assessed the genetic IL6-Receptor variant p.Asp358Ala (rs2228145) which is linked to impaired IL-6 receptor signalling. Homozygous carriers with HFpEF but not HFrEF exhibited significantly lower NT-pro-BNP levels compared with wildtype carriers (HFpEF 779 pg/mL ± 787 vs. 1180 pg/ mL ± 1532; P = 0.008; HFrEF 2289 pg/ mL ± 3439 vs. 2326 pg/ mL ± 3386; P = 0.94), raising the hypothesis that IL-6 signalling may play a pathophysiological role in HFpEF. CONCLUSIONS These data suggest a predictive value of elevated IL-6 for CV-mortality in HFpEF but not in HFrEF patients.
Collapse
Affiliation(s)
- Martin Berger
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| | - Winfried März
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
- Synlab AcademySynlab Holding DeutschlandMannheimGermany
| | - Alexander Niessner
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaViennaAustria
| | - Graciela Delgado
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
| | - Marcus Kleber
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology) Mannheim Medical FacultyUniversity of HeidelbergMannheimGermany
- SYNLAB MVZ Humangenetik MannheimMannheimGermany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Nikolaus Marx
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| | - Katharina Schuett
- Department of Internal Medicine I; University Hospital AachenRWTH UniversityAachenGermany
| |
Collapse
|
10
|
Moissl AP, Delgado GE, Kleber ME, Krämer BK, März W, Lorkowski S. Associations between serum mineral concentrations and mortality by renal function in the Ludwigshafen Risk and Cardiovascular Health Study. Sci Rep 2024; 14:28581. [PMID: 39562674 PMCID: PMC11577029 DOI: 10.1038/s41598-024-79575-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
The association of serum concentrations of minerals and phosphate with overall and cardiovascular mortality based on renal function is poorly understood. 3307 patients (average age 62.7 ± 10.6 years) in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study were grouped by estimated glomerular filtration rate (eGFR) into three categories: < 60, 60-89, and ≥ 90 mL/min per 1.73 m2, per KDIGO 2022 guidelines and were analysed using Cox regression. Low serum sodium and iron concentrations were associated with poor renal function and increased overall mortality risk, whereas higher serum zinc concentrations were associated with reduced overall and cardiovascular mortality risk. Elevated serum copper concentrations were associated with increased mortality risk across all eGFR categories. Comparing low and normal eGFR, we observed a fourfold increase in all-cause mortality risk for eGFR < 60 mL/min per 1.73 m2 and a twofold increase for eGFR 60-89 mL/min per 1.73 m2, accompanied by changes in serum mineral concentrations. The optimal range of mineral and phosphate concentrations in serum was strongly related to renal function. To reduce mortality risk, it's important to regularly monitor serum mineral and phosphate concentrations as well as renal function, especially in cardiovascular patients with compromised renal function.
Collapse
Affiliation(s)
- Angela P Moissl
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Straße 25, 07743, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim, Mannheim, Germany
| | - Bernhard K Krämer
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- European Center for Angioscience (ECAS), Faculty of Medicine, University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland, Augsburg and Mannheim, Mannheim, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Straße 25, 07743, Jena, Germany.
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany.
| |
Collapse
|
11
|
Molnar S, Scharnagl H, Delgado GE, Krämer BK, Laufs U, März W, Kleber ME, Katzmann JL. Clinical and genetic diagnosis of familial hypercholesterolaemia in patients undergoing coronary angiography: the Ludwigshafen Risk and Cardiovascular Health Study. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2024; 10:632-640. [PMID: 38196142 DOI: 10.1093/ehjqcco/qcad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/07/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024]
Abstract
AIMS To investigate the prevalence of familial hypercholesterolaemia (FH) and compare the performance of clinical criteria and genetic testing in patients undergoing coronary angiography. METHODS AND RESULTS The prevalence of FH was determined with the Dutch Lipid Clinical Network (DLCN), US 'Make Early Diagnosis to Prevent Early Death' (US-MEDPED), Simon Broome (SB) criteria, the 'familial hypercholesterolaemia case ascertainment tool' (FAMCAT), and a clinical algorithm. Genetic screening was conducted with a custom array from Affymetrix (CARRENAL array) harbouring 944 FH mutations.The study cohort consisted of 3267 patients [78.6% with coronary artery disease (CAD)]. FH was diagnosed in 2.8%, 2.2%, 3.9%, and 7.9% using the DLCN, US-MEDPED, SB criteria, and the FAMCAT. The clinical algorithm identified the same patients as the SB criteria. Pathogenic FH mutations were found in 1.2% (1.2% in patients with CAD, 1.0% in patients without CAD). FH was more frequently diagnosed in younger patients. With genetic testing as reference, the clinical criteria achieved areas under the ROC curve [area under the curves (AUCs)] in the range of 0.56-0.68. Using only low-density lipoprotein cholesterol (LDL-C) corrected for statin intake, an AUC of 0.68 was achieved. CONCLUSION FH is up to four-fold more prevalent in patients undergoing coronary angiography than in contemporary cohorts representing the general population. Different clinical criteria yield substantially different diagnosis rates, overestimating the prevalence of FH compared with genetic testing. LDL-C testing alone may be sufficient to raise the suspicion of FH, which then needs to be corroborated by genetic testing. LAY SUMMARY In this study, we investigated the frequency of familial hypercholesterolaemia-a common genetic condition leading to markedly elevated low-density lipoprotein (LDL) cholesterol and increased risk of atherosclerosis-in 3267 patients undergoing coronary angiography according to commonly used diagnostic scoring systems and genetic testing.
Collapse
Affiliation(s)
- Stefan Molnar
- Medical Clinic V (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Graciela E Delgado
- Medical Clinic V (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Bernhard K Krämer
- Medical Clinic V (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Ulrich Laufs
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
| | - Winfried März
- Medical Clinic V (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- Synlab Academy, Mannheim, Germany
| | - Marcus E Kleber
- Medical Clinic V (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology, Pneumology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Synlab MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Julius L Katzmann
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
12
|
Delgado GE, Kleber ME, Moissl AP, Winklhofer-Roob BM, Krämer BK, Renner W, Langsenlehner T, Dschietzig TB, März W, Armbruster FP. Haptoglobin polymorphism, vitamin E and mortality: the Ludwigshafen Risk and Cardiovascular Health Study. BMJ Nutr Prev Health 2024; 7:e001061. [PMID: 39882297 PMCID: PMC11773666 DOI: 10.1136/bmjnph-2024-001061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 01/31/2025] Open
Abstract
Objective In humans, haptoglobin (Hp) exists in two allelic forms, Hp1 and Hp2, that differ significantly in their ability to protect the organism from oxidative stress. It has been proposed that in patients with diabetes mellitus carriers of the Hp2-2 genotype may benefit from vitamin E supplementation. Aim of our study was to investigate if there is evidence regarding a potential interaction between the Hp polymorphism and vitamin E with regard to mortality in individuals at medium-to-high cardiovascular risk with and without diabetes mellitus. Research design and methods Data from 3176 participants of the Ludwigshafen Risk and Cardiovascular Health study, a monocentric hospital-based study of patients referred for coronary angiography, were analysed using Cox proportional hazard regression. Results Participants with the Hp2-2 genotype demonstrated significantly lower Hp levels, while carriers of at least one Hp-2 allele displayed elevated levels of the inflammatory markers high-sensitive C reactive protein and serum amyloid A. No notable differences in comorbidities were observed among the various HP genotype groups. While the HP genotype showed no direct association with mortality, a borderline significant correlation between α-tocopherol plasma concentration and overall mortality was noted. An interaction between vitamin E status and the HP genotype regarding mortality risk was evident, particularly among patients with diabetes mellitus, with a p value of 0.021 for the interaction term. In restricted cubic splice analysis, patients with diabetes mellitus who are carriers of the Hp2-2 genotype seem to benefit from higher γ-tocopherol concentrations whereas for the other genotype groups there was a direct association with mortality risk. Conclusion Particularly in patients with diabetes mellitus we could show a significant interaction of γ-tocopherol plasma concentration and HP genotype. Carriers of the Hp2-2 genotype seemed to benefit from higher plasma concentrations of γ-tocopherol. Further research is warranted to elucidate the underlying mechanisms and potential therapeutic implications in cardiovascular disease management.
Collapse
Affiliation(s)
- Graciela E Delgado
- Vth Department of Medicine(Nephrology, Hypertensiology, Rheumatology, Endocrinology, Pneumology), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Marcus E Kleber
- Vth Department of Medicine(Nephrology, Hypertensiology, Rheumatology, Endocrinology, Pneumology), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine(Nephrology, Hypertensiology, Rheumatology, Endocrinology, Pneumology), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | | | - Bernhard K Krämer
- Vth Department of Medicine(Nephrology, Hypertensiology, Rheumatology, Endocrinology, Pneumology), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Wilfried Renner
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University Graz, Graz, Austria
| | | | - Winfried März
- Vth Department of Medicine(Nephrology, Hypertensiology, Rheumatology, Endocrinology, Pneumology), Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
- SYNLAB Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | | |
Collapse
|
13
|
Koloi A, Loukas VS, Hourican C, Sakellarios AI, Quax R, Mishra PP, Lehtimäki T, Raitakari OT, Papaloukas C, Bosch JA, März W, Fotiadis DI. Predicting early-stage coronary artery disease using machine learning and routine clinical biomarkers improved by augmented virtual data. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2024; 5:542-550. [PMID: 39318697 PMCID: PMC11417487 DOI: 10.1093/ehjdh/ztae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/18/2024] [Accepted: 06/01/2024] [Indexed: 09/26/2024]
Abstract
Aims Coronary artery disease (CAD) is a highly prevalent disease with modifiable risk factors. In patients with suspected obstructive CAD, evaluating the pre-test probability model is crucial for diagnosis, although its accuracy remains controversial. Machine learning (ML) predictive models can help clinicians detect CAD early and improve outcomes. This study aimed to identify early-stage CAD using ML in conjunction with a panel of clinical and laboratory tests. Methods and results The study sample included 3316 patients enrolled in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. A comprehensive array of attributes was considered, and an ML pipeline was developed. Subsequently, we utilized five approaches to generating high-quality virtual patient data to improve the performance of the artificial intelligence models. An extension study was carried out using data from the Young Finns Study (YFS) to assess the results' generalizability. Upon applying virtual augmented data, accuracy increased by approximately 5%, from 0.75 to -0.79 for random forests (RFs), and from 0.76 to -0.80 for Gradient Boosting (GB). Sensitivity showed a significant boost for RFs, rising by about 9.4% (0.81-0.89), while GB exhibited a 4.8% increase (0.83-0.87). Specificity showed a significant boost for RFs, rising by ∼24% (from 0.55 to 0.70), while GB exhibited a 37% increase (from 0.51 to 0.74). The extension analysis aligned with the initial study. Conclusion Accurate predictions of angiographic CAD can be obtained using a set of routine laboratory markers, age, sex, and smoking status, holding the potential to limit the need for invasive diagnostic techniques. The extension analysis in the YFS demonstrated the potential of these findings in a younger population, and it confirmed applicability to atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Angela Koloi
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Vasileios S Loukas
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
| | - Cillian Hourican
- Computational Science Lab, Institute of Informatics, University of Amsterdam, Amsterdam, The Netherlands
| | - Antonis I Sakellarios
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
- Biomedical Engineering of the Department of Mechanical Engineering and Aeronautics, University of Patras, Patras, Greece
| | - Rick Quax
- Computational Science Lab, Institute of Informatics, University of Amsterdam, Amsterdam, The Netherlands
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Faculty of Medicine and Health Technology, Finnish Cardiovascular Research Center Tampere, 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
- Faculty of Medicine and Health Technology, Finnish Cardiovascular Research Center Tampere, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Costas Papaloukas
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Jos A Bosch
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Winfried März
- Department of Internal Medicine V, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
- SYNLAB Holding Deutschland GmbH, Augsburg, Germany
| | - Dimitrios I Fotiadis
- Unit of Medical Technology and Intelligent Information Systems, Department of Materials Science and Engineering, University of Ioannina, Ioannina, Greece
- Department of Biomedical Research, FORTH-IMBB, GR 45110 Ioannina, Greece
| |
Collapse
|
14
|
Hörber S, Prystupa K, Jacoby J, Fritsche A, Kleber ME, Moissl AP, Hellstern P, Peter A, März W, Wagner R, Heni M. Blood coagulation in Prediabetes clusters-impact on all-cause mortality in individuals undergoing coronary angiography. Cardiovasc Diabetol 2024; 23:306. [PMID: 39175055 PMCID: PMC11342575 DOI: 10.1186/s12933-024-02402-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Metabolic clusters can stratify subgroups of individuals at risk for type 2 diabetes mellitus and related complications. Since obesity and insulin resistance are closely linked to alterations in hemostasis, we investigated the association between plasmatic coagulation and metabolic clusters including the impact on survival. METHODS Utilizing data from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, we assigned 917 participants without diabetes to prediabetes clusters, using oGTT-derived glucose and insulin, high-density lipoprotein cholesterol, triglycerides, and anthropometric data. We performed a comprehensive analysis of plasmatic coagulation parameters and analyzed their associations with mortality using proportional hazards models. Mediation analysis was performed to assess the effect of coagulation factors on all-cause mortality in prediabetes clusters. RESULTS Prediabetes clusters were assigned using published tools, and grouped into low-risk (clusters 1,2,4; n = 643) and high-risk (clusters 3,5,6; n = 274) clusters. Individuals in the high-risk clusters had a significantly increased risk of death (HR = 1.30; CI: 1.01 to 1.67) and showed significantly elevated levels of procoagulant factors (fibrinogen, FVII/VIII/IX), D-dimers, von-Willebrand factor, and PAI-1, compared to individuals in the low-risk clusters. In proportional hazards models adjusted for relevant confounders, elevated levels of fibrinogen, D-dimers, FVIII, and vWF were found to be associated with an increased risk of death. Multiple mediation analysis indicated that vWF significantly mediates the cluster-specific risk of death. CONCLUSIONS High-risk prediabetes clusters are associated with prothrombotic changes in the coagulation system that likely contribute to the increased mortality in those individuals at cardiometabolic risk. The hypercoagulable state observed in the high-risk clusters indicates an increased risk for cardiovascular and thrombotic diseases that should be considered in future risk stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Sebastian Hörber
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Katsiaryna Prystupa
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Johann Jacoby
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Department for Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Hellstern
- Center of Hemostasis and Thrombosis Zurich, Zurich, Switzerland
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Robert Wagner
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Martin Heni
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
15
|
Pott J, Kheirkhah A, Gadin JR, Kleber ME, Delgado GE, Kirsten H, Forer L, Hauck SM, Burkhardt R, Scharnagl H, Loeffler M, März W, Thiery J, Gieger C, Peters A, Silveira A, Hooft FV, Kronenberg F, Scholz M. Sex and statin-related genetic associations at the PCSK9 gene locus: results of genome-wide association meta-analysis. Biol Sex Differ 2024; 15:26. [PMID: 38532495 PMCID: PMC10964567 DOI: 10.1186/s13293-024-00602-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key player of lipid metabolism with higher plasma levels in women throughout their life. Statin treatment affects PCSK9 levels also showing evidence of sex-differential effects. It remains unclear whether these differences can be explained by genetics. METHODS We performed genome-wide association meta-analyses (GWAS) of PCSK9 levels stratified for sex and statin treatment in six independent studies of Europeans (8936 women/11,080 men respectively 14,825 statin-free/5191 statin-treated individuals). Loci associated in one of the strata were tested for statin- and sex-interactions considering all independent signals per locus. Independent variants at the PCSK9 gene locus were then used in a stratified Mendelian Randomization analysis (cis-MR) of PCSK9 effects on low-density lipoprotein cholesterol (LDL-C) levels to detect differences of causal effects between the subgroups. RESULTS We identified 11 loci associated with PCSK9 in at least one stratified subgroup (p < 1.0 × 10-6), including the PCSK9 gene locus and five other lipid loci: APOB, TM6SF2, FADS1/FADS2, JMJD1C, and HP/HPR. The interaction analysis revealed eight loci with sex- and/or statin-interactions. At the PCSK9 gene locus, there were four independent signals, one with a significant sex-interaction showing stronger effects in men (rs693668). Regarding statin treatment, there were two significant interactions in PCSK9 missense mutations: rs11591147 had stronger effects in statin-free individuals, and rs11583680 had stronger effects in statin-treated individuals. Besides replicating known loci, we detected two novel genome-wide significant associations: one for statin-treated individuals at 6q11.1 (within KHDRBS2) and one for males at 12q24.22 (near KSR2/NOS1), both with significant interactions. In the MR of PCSK9 on LDL-C, we observed significant causal estimates within all subgroups, but significantly stronger causal effects in statin-free subjects compared to statin-treated individuals. CONCLUSIONS We performed the first double-stratified GWAS of PCSK9 levels and identified multiple biologically plausible loci with genetic interaction effects. Our results indicate that the observed sexual dimorphism of PCSK9 and its statin-related interactions have a genetic basis. Significant differences in the causal relationship between PCSK9 and LDL-C suggest sex-specific dosages of PCSK9 inhibitors.
Collapse
Affiliation(s)
- Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK.
| | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jesper R Gadin
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Solna, Sweden
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - 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
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, Synlab Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
- Faculty of Medicine, University of Kiel, Kiel, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Angela Silveira
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Solna, Sweden
| | - Ferdinand Van't Hooft
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital Solna, Solna, Sweden
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany.
- LIFE Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
16
|
Schön M, Prystupa K, Mori T, Zaharia OP, Bódis K, Bombrich M, Möser C, Yurchenko I, Kupriyanova Y, Strassburger K, Bobrov P, Nair ATN, Bönhof GJ, Strom A, Delgado GE, Kaya S, Guthoff R, Stefan N, Birkenfeld AL, Hauner H, Seissler J, Pfeiffer A, Blüher M, Bornstein S, Szendroedi J, Meyhöfer S, Trenkamp S, Burkart V, Schrauwen-Hinderling VB, Kleber ME, Niessner A, Herder C, Kuss O, März W, Pearson ER, Roden M, Wagner R. Analysis of type 2 diabetes heterogeneity with a tree-like representation: insights from the prospective German Diabetes Study and the LURIC cohort. Lancet Diabetes Endocrinol 2024; 12:119-131. [PMID: 38142707 DOI: 10.1016/s2213-8587(23)00329-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Heterogeneity in type 2 diabetes can be represented by a tree-like graph structure by use of reversed graph-embedded dimensionality reduction. We aimed to examine whether this approach can be used to stratify key pathophysiological components and diabetes-related complications during longitudinal follow-up of individuals with recent-onset type 2 diabetes. METHODS For this cohort analysis, 927 participants aged 18-69 years from the German Diabetes Study (GDS) with recent-onset type 2 diabetes were mapped onto a previously developed two-dimensional tree based on nine simple clinical and laboratory variables, residualised for age and sex. Insulin sensitivity was assessed by a hyperinsulinaemic-euglycaemic clamp, insulin secretion was assessed by intravenous glucose tolerance test, hepatic lipid content was assessed by 1 H magnetic resonance spectroscopy, serum interleukin (IL)-6 and IL-18 were assessed by ELISA, and peripheral and autonomic neuropathy were assessed by functional and clinical measures. Participants were followed up for up to 16 years. We also investigated heart failure and all-cause mortality in 794 individuals with type 2 diabetes undergoing invasive coronary diagnostics from the Ludwigshafen Risk and Cardiovascular Health (LURIC) cohort. FINDINGS There were gradients of clamp-measured insulin sensitivity (both dimensions: p<0·0001) and insulin secretion (pdim1<0·0001, pdim2=0·00097) across the tree. Individuals in the region with the lowest insulin sensitivity had the highest hepatic lipid content (n=205, pdim1<0·0001, pdim2=0·037), pro-inflammatory biomarkers (IL-6: n=348, pdim1<0·0001, pdim2=0·013; IL-18: n=350, pdim1<0·0001, pdim2=0·38), and elevated cardiovascular risk (nevents=143, pdim1=0·14, pdim2<0·00081), whereas individuals positioned in the branch with the lowest insulin secretion were more prone to require insulin therapy (nevents=85, pdim1=0·032, pdim2=0·12) and had the highest risk of diabetic sensorimotor polyneuropathy (nevents=184, pdim1=0·012, pdim2=0·044) and cardiac autonomic neuropathy (nevents=118, pdim1=0·0094, pdim2=0·06). In the LURIC cohort, all-cause mortality was highest in the tree branch showing insulin resistance (nevents=488, pdim1=0·12, pdim2=0·0032). Significant gradients differentiated individuals having heart failure with preserved ejection fraction from those who had heart failure with reduced ejection fraction. INTERPRETATION These data define the pathophysiological underpinnings of the tree structure, which has the potential to stratify diabetes-related complications on the basis of routinely available variables and thereby expand the toolbox of precision diabetes diagnosis. FUNDING German Diabetes Center, German Federal Ministry of Health, Ministry of Culture and Science of the state of North Rhine-Westphalia, German Federal Ministry of Education and Research, German Diabetes Association, German Center for Diabetes Research, European Community, German Research Foundation, and Schmutzler Stiftung.
Collapse
Affiliation(s)
- Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Tim Mori
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Oana P Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maria Bombrich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Clara Möser
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Anand T N Nair
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Graciela E Delgado
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Preventive Medicine and Digital Health Baden-Württemberg, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sema Kaya
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rainer Guthoff
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, München, Germany
| | - Jochen Seissler
- Diabetes Research Group, Medical Department 4, Ludwig-Maximilians University Munich, München, Germany
| | - Andreas Pfeiffer
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Matthias Blüher
- Department of Medicine, Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stefan Bornstein
- Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Julia Szendroedi
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Svenja Meyhöfer
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany; Department of Internal Medicine 1, Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Vera B Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Marcus E Kleber
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Austria
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Kuss
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Winfried März
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
| | - Ewan R Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
17
|
Herrmann M, Zelzer S, Cavalier E, Kleber M, Drexler-Helmberg C, Schlenke P, Curcic P, Keppel MH, Enko D, Scharnagl H, Pilz S, März W. Functional Assessment of Vitamin D Status by a Novel Metabolic Approach: The Low Vitamin D Profile Concept. Clin Chem 2023; 69:1307-1316. [PMID: 37798100 DOI: 10.1093/clinchem/hvad151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/24/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Determining serum 25-hydroxyvitamin D [25(OH)D], 24,25-dihydroxyvitamin D [24,25(OH)2D] and the vitamin D metabolite ratio (VMR) allows the identification of individuals with a low vitamin D metabolite profile. Here, we evaluated if such a functional approach provides superior diagnostic information to serum 25(OH)D alone. METHODS 25(OH)D, 24,25(OH)2D, and the VMR were determined in participants of the DESIRE (Desirable Vitamin D Concentrations, n = 2010) and the LURIC (Ludwigshafen Risk and Cardiovascular Health, n = 2456) studies. A low vitamin D metabolite profile (vitamin D insufficiency) was defined by a 24,25(OH)2D concentration <1.2 ng/mL (<3 nmol/L) and a VMR <4%. Parathyroid hormone (PTH) and bone turnover markers were measured in both cohorts, whereas 10-year mortality data was recorded in LURIC only. RESULTS The median age in DESIRE and LURIC was 43.3 and 63.8 years, respectively. Median 25(OH)D concentrations were 27.2 ng/mL (68.0 nmol/L) and 15.5 ng/mL (38.8 nmol/L), respectively. Serum 25(OH)D deficiency, defined as <20.2 ng/mL (<50 nmol/L), was present in 483 (24.0%) and 1701 (69.3%) participants of DESIRE and LURIC, respectively. In contrast, only 77 (3.8%) and 521 (21.2%) participants had a low vitamin D metabolite profile. Regardless of the serum 25(OH)D concentration, a low vitamin D metabolite profile was associated with a significantly higher PTH, accelerated bone metabolism, and higher all-cause mortality than an unremarkable vitamin D metabolite profile. CONCLUSIONS The personalized assessment of vitamin D status using a functional approach better identifies patients with accelerated bone metabolism and increased mortality than the use of a fixed 25(OH)D cutoff of 20 ng/mL (50 nmol/L).
Collapse
Affiliation(s)
- Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sieglinde Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liege, Liege, Belgium
| | - Marcus Kleber
- Department of Internal Medicine 5 (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
- Synlab Human Genetics Laboratory, Synlab AG, Mannheim, Germany
| | - Camilla Drexler-Helmberg
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Peter Schlenke
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Pero Curcic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Martin H Keppel
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Dietmar Enko
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Stefan Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Human Genetics Laboratory, Synlab AG, Mannheim, Germany
- Synlab Academy, Synlab Holding Germany GmbH, Mannheim, Germany
| |
Collapse
|
18
|
Zheng J, Wheeler E, Pietzner M, Andlauer TFM, Yau MS, Hartley AE, Brumpton BM, Rasheed H, Kemp JP, Frysz M, Robinson J, Reppe S, Prijatelj V, Gautvik KM, Falk L, Maerz W, Gergei I, Peyser PA, Kavousi M, de Vries PS, Miller CL, Bos M, van der Laan SW, Malhotra R, Herrmann M, Scharnagl H, Kleber M, Dedoussis G, Zeggini E, Nethander M, Ohlsson C, Lorentzon M, Wareham N, Langenberg C, Holmes MV, Davey Smith G, Tobias JH. Lowering of Circulating Sclerostin May Increase Risk of Atherosclerosis and Its Risk Factors: Evidence From a Genome-Wide Association Meta-Analysis Followed by Mendelian Randomization. Arthritis Rheumatol 2023; 75:1781-1792. [PMID: 37096546 PMCID: PMC10586470 DOI: 10.1002/art.42538] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 03/22/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE In this study, we aimed to establish the causal effects of lowering sclerostin, target of the antiosteoporosis drug romosozumab, on atherosclerosis and its risk factors. METHODS A genome-wide association study meta-analysis was performed of circulating sclerostin levels in 33,961 European individuals. Mendelian randomization (MR) was used to predict the causal effects of sclerostin lowering on 15 atherosclerosis-related diseases and risk factors. RESULTS We found that 18 conditionally independent variants were associated with circulating sclerostin. Of these, 1 cis signal in SOST and 3 trans signals in B4GALNT3, RIN3, and SERPINA1 regions showed directionally opposite signals for sclerostin levels and estimated bone mineral density. Variants with these 4 regions were selected as genetic instruments. MR using 5 correlated cis-SNPs suggested that lower sclerostin increased the risk of type 2 diabetes mellitus (DM) (odds ratio [OR] 1.32 [95% confidence interval (95% CI) 1.03-1.69]) and myocardial infarction (MI) (OR 1.35 [95% CI 1.01-1.79]); sclerostin lowering was also suggested to increase the extent of coronary artery calcification (CAC) (β = 0.24 [95% CI 0.02-0.45]). MR using both cis and trans instruments suggested that lower sclerostin increased hypertension risk (OR 1.09 [95% CI 1.04-1.15]), but otherwise had attenuated effects. CONCLUSION This study provides genetic evidence to suggest that lower levels of sclerostin may increase the risk of hypertension, type 2 DM, MI, and the extent of CAC. Taken together, these findings underscore the requirement for strategies to mitigate potential adverse effects of romosozumab treatment on atherosclerosis and its related risk factors.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of BristolBristolUK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeUK
| | - Maik Pietzner
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK, and Computational Medicine, Berlin Institute of Health at Charité–Universitätsmedizin BerlinBerlinGermany
| | - Till F. M. Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
| | - Michelle S. Yau
- Marcus Institute for Aging Research, Hebrew SeniorLifeHarvard Medical SchoolBostonMassachusetts
| | | | - Ben Michael Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, and HUNT Research Centre, Department of Public Health and Nursing, NTNUNorwegian University of Science and TechnologyLevangerNorway
| | - Humaira Rasheed
- MRC IEU, Bristol Medical School, University of Bristol, Bristol, UK, and HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway, and Division of Medicine and Laboratory Sciences, Faculty of MedicineUniversity of OsloOsloNorway
| | - John P. Kemp
- MRC IEU, Bristol Medical School, University of Bristol, Bristol, UK, and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia, and The University of Queensland Diamantina InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | - Monika Frysz
- MRC IEU, Bristol Medical School, University of Bristol, and Musculoskeletal Research UnitUniversity of BristolBristolUK
| | - Jamie Robinson
- MRC IEU, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Sjur Reppe
- Unger‐Vetlesen Institute, Lovisenberg Diaconal Hospital and Department of Plastic and Reconstructive Surgery, Oslo University Hospital and Department of Medical BiochemistryOslo University HospitalOsloNorway
| | - Vid Prijatelj
- Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| | | | - Louise Falk
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK, and Computational Medicine, Berlin Institute of Health at Charité–Universitätsmedizin BerlinBerlinGermany
| | - Winfried Maerz
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria, and SYNLAB Academy, SYNLAB Holding Deutschland GmbH and Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty MannheimUniversity of HeidelbergMannheimGermany
| | - Ingrid Gergei
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, and Therapeutic Area Cardiovascular MedicineBoehringer Ingelheim International GmbHIngelheimGermany
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public HealthUniversity of MichiganAnn Arbor
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public HealthThe University of Texas Health Science Center at Houston
| | - Clint L. Miller
- Center for Public Health Genomics, Department of Public Health SciencesUniversity of VirginiaCharlottesville
| | - Maxime Bos
- Department of Epidemiology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division of Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Rajeev Malhotra
- Cardiology Division, Department of MedicineMassachusetts General HospitalBoston
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria
| | - Marcus Kleber
- SYNLAB Academy, SYNLAB Holding Deutschland GmbHMannheimGermany
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and EducationHarokopio UniversityAthensGreece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, and Technical University of Munich (TUM) and Klinikum Rechts der IsarTUM School of MedicineMunichGermany
| | - Maria Nethander
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg and Bioinformatics and Data Centre, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of MedicineUniversity of GothenburgGothenburgSweden
| | - Mattias Lorentzon
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and Region Västra Götaland, Geriatric Medicine, Sahlgrenska University Hospital, Mölndal, Sweden, and Mary McKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneVictoriaAustralia
| | - Nick Wareham
- MRC Epidemiology Unit, Institute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeUK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK, and Computational Medicine, Berlin Institute of Health at Charité–Universitätsmedizin BerlinBerlinGermany
| | - Michael V. Holmes
- MRC IEU, Bristol Medical School, University of Bristol, and Medical Research Council Population Health Research Unit, University of Oxford, and Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population HealthUniversity of Oxford, and National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University HospitalOxfordUK
| | | | - Jonathan H. Tobias
- MRC IEU, Bristol Medical School, University of Bristol, and Musculoskeletal Research UnitUniversity of BristolBristolUK
| |
Collapse
|
19
|
Prystupa K, Delgado GE, Moissl AP, Kleber ME, Birkenfeld AL, Heni M, Fritsche A, März W, Wagner R. Clusters of prediabetes and type 2 diabetes stratify all-cause mortality in a cohort of participants undergoing invasive coronary diagnostics. Cardiovasc Diabetol 2023; 22:211. [PMID: 37592260 PMCID: PMC10436494 DOI: 10.1186/s12933-023-01923-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Heterogeneous metabolic clusters have been identified in diabetic and prediabetic states. It is not known whether such pathophysiologic clusters impact survival in at-risk persons being evaluated for coronary heart disease. METHODS The LURIC Study recruited patients referred for coronary angiography at a median age of 63 (IQR 56-70) with a follow-up of 16.1 (IQR 9.6, 17.7) years. Clustering of 1269 subjects without diabetes was performed with oGTT-derived glucose and insulin; fasting triglyceride, high-density lipoprotein, BMI, waist and hip circumference. Patients with T2D (n = 794) were clustered using age, BMI, glycemia, homeostasis model assessment, and islet autoantibodies. Associations of clusters with mortality were analysed using Cox regression. RESULTS Individuals without diabetes were classified into six subphenotypes, with 884 assigned to subjects at low-risk (cluster 1,2,4) and 385 at high-risk (cluster 3,5,6) for diabetes. We found significantly increased mortality in clusters 3 (hazard ratio (HR)1.42), 5 (HR 1.43), and 6 (HR 1.46) after adjusting for age, BMI, HbA1c and sex. In the T2D group, 508 were assigned to mild age-related diabetes (MARD), 183 to severe insulin-resistant diabetes (SIRD), 84 to mild obesity-related diabetes (MOD), 19 to severe insulin-deficient diabetes (SIDD). Compared to the low-risk non-diabetes group, crude mortality was not different in MOD. Increased mortality was found for MARD (HR 2.2), SIRD (HR 2.2), and SIDD (HR 2.5). CONCLUSIONS Metabolic clustering successfully stratifies survival even among persons undergoing invasive coronary diagnostics. Novel clustering approaches based on glucose metabolism can identify persons who require special attention as they are at risk of increased mortality.
Collapse
Affiliation(s)
- Katsiaryna Prystupa
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| | - Andreas Fritsche
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
| | - Robert Wagner
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
| |
Collapse
|
20
|
Krämer RM, Moissl AP, Lorkowski S, Krämer BK, Lehtimäki T, Mishra BH, Mishra PP, Leipe J, März W, Kleber ME, Müller-Myhsok B, Delgado GE. High genetic risk for depression as an independent risk factor for mortality in patients referred for coronary angiography. Front Cardiovasc Med 2023; 10:1125151. [PMID: 37435051 PMCID: PMC10330785 DOI: 10.3389/fcvm.2023.1125151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/08/2023] [Indexed: 07/13/2023] Open
Abstract
Background Different observations have suggested that patients with depression have a higher risk for a number of comorbidities and mortality. The underlying causes have not been fully understood yet. Aims The aim of our study was to investigate the association of a genetic depression risk score (GDRS) with mortality [all-cause and cardiovascular (CV)] and markers of depression (including intake of antidepressants and a history of depression) in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study involving 3,316 patients who had been referred for coronary angiography. Methods and results The GDRS was calculated in 3,061 LURIC participants according to a previously published method and was found to be associated with all-cause (p = 0.016) and CV mortality (p = 0.0023). In Cox regression models adjusted for age, sex, body mass index, LDL-cholesterol, HDL-cholesterol, triglycerides, hypertension, smoking, and diabetes mellitus, the GDRS remained significantly associated with all-cause [1.18 (1.04-1.34, p = 0.013)] and CV [1.31 (1.11-1.55, p = 0.001)] mortality. The GDRS was not associated with the intake of antidepressants or a history of depression. However, this cohort of CV patients had not specifically been assessed for depression, leading to marked underreporting. We were unable to identify any specific biomarkers correlated with the GDRS in LURIC participants. Conclusion A genetic predisposition for depression estimated by a GDRS was independently associated with all-cause and CV mortality in our cohort of patients who had been referred for coronary angiography. No biomarker correlating with the GDRS could be identified.
Collapse
Affiliation(s)
- Robert M. Krämer
- Department for Children and Adolescent Psychiatry, Central Institute for Mental Health Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Angela P. Moissl
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Bernhard K. Krämer
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience ECAS, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Binisha H. Mishra
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, 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, Tampere, Finland
| | - Jan Leipe
- 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
| | - Marcus E. Kleber
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- European Center for Angioscience ECAS, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute for Psychiatry, Munich, Germany
- Department of Health Data Science, University of Liverpool, Liverpool, United Kingdom
| | - Graciela E. Delgado
- Vth Department of Medicine, University Medicine Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
21
|
Kovilakath A, Wohlford G, Cowart LA. Circulating sphingolipids in heart failure. Front Cardiovasc Med 2023; 10:1154447. [PMID: 37229233 PMCID: PMC10203217 DOI: 10.3389/fcvm.2023.1154447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
Lack of significant advancements in early detection and treatment of heart failure have precipitated the need for discovery of novel biomarkers and therapeutic targets. Over the past decade, circulating sphingolipids have elicited promising results as biomarkers that premonish adverse cardiac events. Additionally, compelling evidence directly ties sphingolipids to these events in patients with incident heart failure. This review aims to summarize the current literature on circulating sphingolipids in both human cohorts and animal models of heart failure. The goal is to provide direction and focus for future mechanistic studies in heart failure, as well as pave the way for the development of new sphingolipid biomarkers.
Collapse
Affiliation(s)
- Anna Kovilakath
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - George Wohlford
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, United States
| | - L. Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Richmond Veteran's Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
22
|
Moissl AP, Lorkowski S, Meinitzer A, Pilz S, Scharnagl H, Delgado GE, Kleber ME, Krämer BK, Pieske B, Grübler MR, Brussee H, von Lewinski D, Toplak H, Fahrleitner-Pammer A, März W, Tomaschitz A. Association of branched-chain amino acids with mortality-the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. iScience 2023; 26:106459. [PMID: 37020954 PMCID: PMC10067756 DOI: 10.1016/j.isci.2023.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
Branched-chain amino acids (BCAAs) are effectors of metabolic diseases, but their impact on mortality is largely unknown. We investigated the association of BCAA with risk factors and mortality in 2,236 participants of the Ludwigshafen Risk and Cardiovascular Health (LURIC) study using linear and Cox regression. Adiponectin, hemoglobin, C-peptide, hemoglobin A1c, and homoarginine showed the strongest association with BCAA concentration (all p < 0.001). During a median follow-up of 10.5 years, 715 participants died, including 450 cardiovascular-related deaths. BCAA concentrations were inversely associated with the risk of all-cause and cardiovascular mortality (HR [95% CI] per 1-SD increase in log-BCAA: 0.75 [0.69-0.82] and 0.72 [0.65-0.80], respectively) after adjustment for potential confounders. BCAAs are directly associated with metabolic risk but inversely with mortality in persons with intermediate-to-high cardiovascular risk. Further studies are warranted to evaluate the diagnostic and therapeutic utility of BCAA in the context of cardiovascular diseases.
Collapse
|
23
|
Platelet Reactivity and Cardiovascular Mortality Risk in the LURIC Study. J Clin Med 2023; 12:jcm12051913. [PMID: 36902699 PMCID: PMC10003439 DOI: 10.3390/jcm12051913] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/14/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND The clinical and prognostic implications of platelet reactivity (PR) testing in a P2Y12-inhibitor naïve population are poorly understood. OBJECTIVES This explorative study aims to assess the role of PR and explore factors that may modify elevated mortality risk in patients with altered PR. METHODS Platelet ADP-induced CD62P and CD63 expression were measured by flow-cytometry in 1520 patients who were referred for coronary angiography in the Ludwigshafen Risk and Cardiovascular Health Study (LURIC). RESULTS High- and Low-platelet reactivity to ADP were strong predictors of cardiovascular and all-cause mortality and risk equivalent to the presence of coronary artery disease. (High platelet reactivity 1.4 [95% CI 1.1-1.9]; Low platelet reactivity: 1.4 [95% CI 1.0-2.0]). Relative weight analysis indicated glucose control (HbA1c), renal function ([eGFR]), inflammation (high-sensitive C-reactive protein [hsCRP]) and antiplatelet therapy by Aspirin as consistent mortality risk modifiers in patients with Low- and High-platelet reactivity. Pre-specified stratification of patients by risk modifiers HbA1c (<7.0%), eGFR (>60 mL/min/1.73 m2) and CRP (<3 mg/L) was associated with a lower mortality risk, however irrespective of platelet reactivity. Aspirin treatment was associated with reduced mortality in patients with high platelet reactivity only (p for interaction: 0.02 for CV-death [<0.01 for all-cause mortality]. CONCLUSIONS Cardiovascular mortality risk in patients with High- and Low platelet reactivity is equivalent to the presence of coronary artery disease. Targeted glucose control, improved kidney function and lower inflammation are associated with reduced mortality risk, however independent of platelet reactivity. In contrast, only in patients with High-platelet reactivity was Aspirin treatment associated with lower mortality.
Collapse
|
24
|
Galectin-3 and Blood Group: Binding Properties, Effects on Plasma Levels, and Consequences for Prognostic Performance. Int J Mol Sci 2023; 24:ijms24054415. [PMID: 36901846 PMCID: PMC10002292 DOI: 10.3390/ijms24054415] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Previous studies have reported an association between ABO type blood group and cardiovascular (CV) events and outcomes. The precise mechanisms underpinning this striking observation remain unknown, although differences in von Willebrand factor (VWF) plasma levels have been proposed as an explanation. Recently, galectin-3 was identified as an endogenous ligand of VWF and red blood cells (RBCs) and, therefore, we aimed to explore the role of galectin-3 in different blood groups. Two in vitro assays were used to assess the binding capacity of galectin-3 to RBCs and VWF in different blood groups. Additionally, plasma levels of galectin-3 were measured in different blood groups in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study (2571 patients hospitalized for coronary angiography) and validated in a community-based cohort of the Prevention of Renal and Vascular End-stage Disease (PREVEND) study (3552 participants). To determine the prognostic value of galectin-3 in different blood groups, logistic regression and cox regression models were used with all-cause mortality as the primary outcome. First, we demonstrated that galectin-3 has a higher binding capacity for RBCs and VWF in non-O blood groups, compared to blood group O. Additionally, LURIC patients with non-O blood groups had substantially lower plasma levels of galectin-3 (15.0, 14.9, and 14.0 μg/L in blood groups A, B, and AB, respectively, compared to 17.1 μg/L in blood group O, p < 0.0001). Finally, the independent prognostic value of galectin-3 for all-cause mortality showed a non-significant trend towards higher mortality in non-O blood groups. Although plasma galectin-3 levels are lower in non-O blood groups, the prognostic value of galectin-3 is also present in subjects with a non-O blood group. We conclude that physical interaction between galectin-3 and blood group epitopes may modulate galectin-3, which may affect its performance as a biomarker and its biological activity.
Collapse
|
25
|
Zhang W, Jin JL, Zhang HW, Zhu YX, Dong Q, Sun J, Guo YL, Dou KF, Xu RX, Li JJ. The value of HDL subfractions in predicting cardiovascular outcomes in untreated, diabetic patients with stable coronary artery disease: An age- and gender-matched case-control study. Front Endocrinol (Lausanne) 2023; 13:1041555. [PMID: 36714594 PMCID: PMC9877453 DOI: 10.3389/fendo.2022.1041555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE The aim of the present study was to examine the value of high-density lipoprotein (HDL) subfractions for predicting cardiovascular events (CVEs) in untreated type 2 diabetes mellitus (T2DM) patients with stable coronary artery disease (SCAD) using an age- and gender-matched case-control study. METHODS In total, 185 SCAD patients and 185 T2DM patients with SCAD were enrolled and subjected to a clinical follow-up of CVEs. HDL subfractions were analyzed using the Quantimetrix Lipoprint System. The relationship between HDL subfractions and CVEs in T2DM patients with SCAD was evaluated by Kaplan-Meier analysis and Cox proportional hazard models. RESULTS During a median 37.7-month follow-up, T2DM patients with SCAD had a higher percentage of CVEs compared to SCAD patients (p=0.039). The concentration of the combined intermediate and small HDL-C subfraction (defined as the mixed HDL subfraction) was related to the event incidence in T2DM patients with SCAD (p=0.004), and it was positively associated with increased CVEs even after adjustment in three models. Kaplan-Meier curve analysis indicated that T2DM patients with SCAD in the high mixed HDL subfraction group (>28 mg/dL) had lower event-free survival rates (p=0.008). CONCLUSIONS Elevated concentration of the mixed HDL subfraction concentration predicts events in T2DM patients with SCAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rui-Xia Xu
- State Key Laboratory of Cardiovascular Disease, Cardiometabolic Medicine Center, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- State Key Laboratory of Cardiovascular Disease, Cardiometabolic Medicine Center, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Yazdani B, Delgado GE, Kleber ME, Yücel G, Husain‐Syed F, Kraemer TD, Jochims J, Leipe J, März W, Krämer BK. The renin-angiotensin-aldosterone system, neurohumoral axis and cardiovascular mortality in LURIC. J Clin Hypertens (Greenwich) 2022; 24:1587-1597. [PMID: 36349861 PMCID: PMC9731599 DOI: 10.1111/jch.14593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/18/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022]
Abstract
Although neurohormones and Renin-Angiotensin-Aldosterone-System (RAAS) components are important predictors of cardiovascular mortality (CVM), their importance for predicting outcomes in patients with/without RAAS-blockers and different degrees of arterial stiffness is less understood. We therefore analyzed long-term data from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study in 3316 patients subdivided according to pulse pressure (PP) and RAAS-blocker use. Patients on RAAS-inhibition had higher renin and noradrenaline, lower aldosterone and aldosterone/renin quotient (ARQ). Renin and noradrenaline significantly predicted CVM in patients without RAAS-blocker (HR = 1.17, 1.15) and in patients receiving angiotensin-converting-enzyme (ACE) inhibitors (HR = 1.17, 1.29), whereas aldosterone predicted CVM only in patients receiving ACE-inhibitors (HR = 1.13). CVM was predicted independently from PP by renin, noradrenaline and angiotensin II. Independently from RAAS inhibition renin decreased and ARQs increased with rising PP. Furthermore, noradrenaline increased with PP, but only without ACE-inhibition. The HR for CVM in the ACE-inhibitor group were 1.29, 1.28, 1.29 for renin in the first, second and third PP quartiles and 1.22, and 1.19 for aldosterone in the second and fourth quartile. Furthermore, we showed that noradrenaline predicts CVM in all PP quartiles in patients with ACE-inhibition. In the RAAS-blocker-free group, the HR for renin for CVM were 1.36 and 1.18 in the third and fourth PP quartiles, but neither aldosterone nor noradrenaline were predictive for CVM within the PP quartiles. Renin and noradrenaline are strong predictors of CVM regardless of RAAS blockade, whereas aldosterone is predictive only in the ACE-inhibitor group. Catecholamines but not renin are associated with rising PP.
Collapse
Affiliation(s)
- Babak Yazdani
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany
| | - Graciela E. Delgado
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany,Center for Preventive Medicine and Digital Health Baden‐Württemberg (CPDBW)Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Marcus E. Kleber
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany,SYNLAB MVZ Humangenetik MannheimMannheimGermany
| | - Gökhan Yücel
- First Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany
| | - Faeq Husain‐Syed
- Second Department of MedicineUniversity Medical Center Giessen UKGMGiessenGermany
| | - Thomas D. Kraemer
- Department of Nephrology and HypertensionHannover Medical SchoolHanoverGermany
| | - Jan Jochims
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany
| | - Jan Leipe
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany
| | - Winfried März
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany,Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University of GrazGrazAustria,Synlab AcademySYNLAB Holding Deutschland GmbHMannheim and AugsburgGermany
| | - Bernhard K. Krämer
- Fifth Department of MedicineUniversity Medical Center Mannheim UMMFaculty of Medicine of the University of HeidelbergMannheimGermany,Center for Preventive Medicine and Digital Health Baden‐Württemberg (CPDBW)Medical Faculty MannheimHeidelberg UniversityMannheimGermany,European Center for Angioscience ECASFaculty of Medicine of the University of HeidelbergMannheimGermany
| |
Collapse
|
27
|
Marttila S, Tamminen H, Rajić S, Mishra PP, Lehtimäki T, Raitakari O, Kähönen M, Kananen L, Jylhävä J, Hägg S, Delerue T, Peters A, Waldenberger M, Kleber ME, März W, Luoto R, Raitanen J, Sillanpää E, Laakkonen EK, Heikkinen A, Ollikainen M, Raitoharju E. Methylation status of VTRNA2-1/ nc886 is stable across populations, monozygotic twin pairs and in majority of tissues. Epigenomics 2022; 14:1105-1124. [PMID: 36200237 DOI: 10.2217/epi-2022-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims & methods: The aim of this study was to characterize the methylation level of a polymorphically imprinted gene, VTRNA2-1/nc886, in human populations and somatic tissues.48 datasets, consisting of more than 30 tissues and >30,000 individuals, were used. Results: nc886 methylation status is associated with twin status and ethnic background, but the variation between populations is limited. Monozygotic twin pairs present concordant methylation, whereas ∼30% of dizygotic twin pairs present discordant methylation in the nc886 locus. The methylation levels of nc886 are uniform across somatic tissues, except in cerebellum and skeletal muscle. Conclusion: The nc886 imprint may be established in the oocyte, and, after implantation, the methylation status is stable, excluding a few specific tissues.
Collapse
Affiliation(s)
- Saara Marttila
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Gerontology Research Center, Tampere University, Tampere, 33014, Finland
| | - Hely Tamminen
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Sonja Rajić
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Fimlab Laboratories, Arvo Ylpön katu 4, Tampere, 33520, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Fimlab Laboratories, Arvo Ylpön katu 4, Tampere, 33520, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku & Turku University Hospital, Turku, 20014, Finland.,Research Centre of Applied & Preventive Cardiovascular Medicine, University of Turku, Turku, 20014, Finland.,Department of Clinical Physiology & Nuclear Medicine, Turku University Hospital, Turku, 20014, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Department of Clinical Physiology, Tampere University Hospital, Tampere, 33521, Finland
| | - Laura Kananen
- Faculty of Medicine & Health Technology, & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520,Finland.,Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden.,Faculty of Social Sciences (Health Sciences), & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Juulia Jylhävä
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden.,Faculty of Social Sciences (Health Sciences), & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Sara Hägg
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Thomas Delerue
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764,, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764,, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,Competence Cluster for Nutrition & Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, 07743, Germany.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg, 86156, Germany.,Clinical Institute of Medical & Chemical Laboratory Diagnostics, Medical University of Graz, Graz, 8010, Austria
| | - Riitta Luoto
- The Social Insurance Institute of Finland (Kela), Helsinki, 00250, Finland.,The UKK Institute for Health Promotion Research, Kaupinpuistonkatu 1, Tampere, 33500, Finland
| | - Jani Raitanen
- The UKK Institute for Health Promotion Research, Kaupinpuistonkatu 1, Tampere, 33500, Finland.,Faculty of Social Sciences (Health Sciences), Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Elina Sillanpää
- Gerontology Research Center & Faculty of Sport & Health Sciences, University of Jyväskylä, Jyväskylä, 40014, Finland.,Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Eija K Laakkonen
- Gerontology Research Center & Faculty of Sport & Health Sciences, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Emma Raitoharju
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| |
Collapse
|
28
|
Biomarkers for Non-Invasive Stratification of Coronary Artery Disease and Prognostic Impact on Long-Term Survival in Patients with Stable Coronary Heart Disease. Nutrients 2022; 14:nu14163433. [PMID: 36014939 PMCID: PMC9413764 DOI: 10.3390/nu14163433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Knowledge about cardiac and inflammatory biomarkers in patients with stable coronary artery disease (CAD) is limited. To address this, we analyzed 3072 patients (36% female) with a median follow-up of 10 years in the Leipzig LIFE Heart Study with suspected CAD with coronary angiography. Selected biomarkers included troponin T (hsTNT), N-terminal pro B-type natriuretic peptide (NT-proBNP), copeptin, C-reactive protein (hsCRP), and interleukin-6 (IL-6). Patients were stratified by CAD severity: CAD0 (no sclerosis), CAD1 (non-obstructive, i.e., stenosis < 50%), and CAD2 (≥one stenosis ≥ 50%). Group comparison (GC) included GC1: CAD0 + 1 vs. CAD2; GC2: CAD0 vs. CAD1 + 2. CAD0, CAD1, and CAD2 were apparent in 1271, 631, and 1170 patients, respectively. Adjusted for classical risk factors, hs-cTnT, NT-proBNP, and IL-6 differed significantly in both GC and hsCRP only in GC2. After multivariate analysis, hs-cTnT, NT-proBNP, and IL-6 remained significant in GC1. In GC2, hs-cTnT (p < 0.001) and copeptin (p = 0.014) reached significance. Ten-year survival in groups CAD0, CAD1, and CAD2 was 88.3%, 77.3%, and 72.4%. Incorporation of hs-cTnT, NT-proBNP, copeptin, and IL-6 improved risk prediction (p < 0.001). The studied cardiac and inflammatory biomarkers enable fast and precise non-invasive identification of mortality risk in CAD patients, allowing the tailoring of primary and secondary CAD prevention.
Collapse
|
29
|
Cruz V, Triviño J, ARANCIBIA VERONICA. Development of electroanalytical methods using different electrodes for the determination of Progesterone (P4) in pharmaceutical formulations and human, cow and goat milk. ELECTROANAL 2022. [DOI: 10.1002/elan.202200272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Ritsch A, Hunjadi M, Stojakovic T, Scherberich JE, Silbernagel G, Scharnagl H, Delgado GE, Kleber ME, März W. Independent Effects of Kidney Function and Cholesterol Efflux on Cardiovascular Mortality. Biomedicines 2022; 10:1832. [PMID: 36009383 PMCID: PMC9404976 DOI: 10.3390/biomedicines10081832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Impaired renal function is associated with cardiovascular and all-cause mortality. In the general population, HDL-cholesterol is associated with cardiovascular events, which is not true in patients with chronic kidney disease (CKD). This has been attributed to abnormal HDL function in CKD. METHODS In this study, we analyzed the association of genetic markers for kidney function with cholesterol efflux capacity as one of the major HDL functions, as well as with cardiovascular mortality, in 2469 patients of the Ludwigshafen Risk and Cardiovascular Health Study who all underwent coronary angiography. RESULTS A genetic score of 53 SNPs associated with GRF and the uromodulin SNP rs12917707 were inversely correlated with cholesterol efflux capacity. This was in line with the observed association between cholesterol efflux capacity and kidney function in these patients. Adjustment for eGFR and uromodulin as markers of kidney function did not affect the relationship between cholesterol efflux and cardiovascular mortality. CONCLUSIONS Our data propose the view that cholesterol efflux and kidney function are exerting their effects on cardiovascular mortality via different and independent pathways. Decreased cholesterol efflux may therefore not mediate the effects of impaired kidney function on cardiovascular mortality.
Collapse
Affiliation(s)
- Andreas Ritsch
- Department of Internal Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Monika Hunjadi
- Department of Internal Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, 8036 Graz, Austria;
| | - Jürgen E. Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum Muenchen-Harlaching, Teaching Hospital of the Ludwig-Maximilians University, 81545 Munich, Germany;
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (H.S.); (W.M.)
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (G.E.D.); (M.E.K.)
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (G.E.D.); (M.E.K.)
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria; (H.S.); (W.M.)
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (G.E.D.); (M.E.K.)
- Synlab Academy, Synlab Holding Deutschland GmbH, 86156 Augsburg, Germany
| |
Collapse
|
31
|
Tsarapatsani K, Sakellarios AI, Pezoulas VC, Tsakanikas VD, Kleber ME, Marz W, Michalis LK, Fotiadis DI. Machine Learning Models for Cardiovascular Disease Events Prediction. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:1066-1069. [PMID: 36085658 DOI: 10.1109/embc48229.2022.9871121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cardiovascular diseases (CVDs) are among the most serious disorders leading to high mortality rates worldwide. CVDs can be diagnosed and prevented early by identifying risk biomarkers using statistical and machine learning (ML) models, In this work, we utilize clinical CVD risk factors and biochemical data using machine learning models such as Logistic Regression (LR), Support Vector Machine (SVM), Random Forest (RF), Naïve Bayes (NB), Extreme Grading Boosting (XGB) and Adaptive Boosting (AdaBoost) to predict death caused by CVD within ten years of follow-up. We used the cohort of the Ludwigshafen Risk and Cardiovascular Health (LURIC) study and 2943 patients were included in the analysis (484 annotated as dead due to CVD). We calculated the Accuracy (ACC), Precision, Recall, F1-Score, Specificity (SPE) and area under the receiver operating characteristic curve (AUC) of each model. The findings of the comparative analysis show that Logistic Regression has been proven to be the most reliable algorithm having accuracy 72.20 %. These results will be used in the TIMELY study to estimate the risk score and mortality of CVD in patients with 10-year risk.
Collapse
|
32
|
Yazdani B, Kleber ME, Yücel G, Delgado GE, Husain-Syed F, Krüger B, März W, Schwenke K, Sigl M, Krämer BK. Polyvascular disease, pulse pressure and mortality. VASA 2022; 51:229-238. [PMID: 35603601 DOI: 10.1024/0301-1526/a001011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Peripheral arterial disease (PAD), coronary artery disease (CAD) and carotid stenosis (CS) are robust predictors of mortality. The value of individual vascular beds in polyvascular disease (PVD) to predict mortality in patients with atherosclerotic burden is not clear. Therefore, we have examined the predictive value of PAD, CAD and CS in patients at intermediate to high risk of cardiovascular (CV) disease. Patients and methods: In our retrospective observational study we analyzed baseline data from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, a monocentric cohort study of 3316 patients referred to coronary angiography. Results: As the number of atherosclerotic vascular beds increased, the hazard ratios (HRs) for both all-cause mortality and CV mortality significantly increased in a multivariate analysis after adjusting for age, sex, body mass index, diabetes mellitus and estimated glomerular filtration rate, with HRs of 1.36 (95%CI: 1.11-1.68), 2.56 (95%CI: 2.01-3.26), 2.84 (95%CI: 1.93-4.17) and 1.56 (95%CI: 1.19-2.06), 2.70 (95%CI: 1.97-3.72), 3.50 (95%CI: 2.19-5.62), respectively. The combination of PAD with either CAD or CS was associated with higher HRs for all-cause (HR 2.81 and 7.53, respectively) and CV (HRs 2.80 and 6.03, respectively) mortality compared with the combination of CAD and CS (HRs 1.94 and 2.43, respectively). The presence of PVD was associated with higher age, systolic blood pressure, pulse pressure (PP; a marker of vascular stiffness), former smoking and inversely with lower eGFR. Conclusions: We show that as the number of atherosclerotic vascular beds increases, all-cause and CV mortality rates increase in parallel. Simultaneous prevalence of PAD is associated with significantly higher all-cause and CV mortality rates compared with CS coexistence. Furthermore, increasing atherosclerotic load may contribute to vascular stiffness and impaired renal function.
Collapse
Affiliation(s)
- Babak Yazdani
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,SYNLAB MVZ Humangenetik Mannheim, Germany
| | - Gökhan Yücel
- First Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany
| | - Graciela E Delgado
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Faeq Husain-Syed
- Department of Internal Medicine II, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Germany
| | - Bernd Krüger
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany
| | - Winfried März
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria.,Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Kay Schwenke
- Division of Vascular Surgery, Department of Surgery, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,The authors contributed equally as senior authors
| | - Martin Sigl
- First Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,The authors contributed equally as senior authors
| | - Bernhard K Krämer
- Fifth Department of Medicine, Faculty of Medicine of the University of Heidelberg, University Medical Center Mannheim UMM, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience ECAS, Faculty of Medicine of the University of Heidelberg, Mannheim, Germany.,The authors contributed equally as senior authors
| |
Collapse
|
33
|
The LDL Apolipoprotein B-to-LDL Cholesterol Ratio: Association with Cardiovascular Mortality and a Biomarker of Small, Dense LDLs. Biomedicines 2022; 10:biomedicines10061302. [PMID: 35740324 PMCID: PMC9220033 DOI: 10.3390/biomedicines10061302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022] Open
Abstract
Background and Objective: Small, dense low-density lipoproteins (LDLs) are considered more atherogenic than normal size LDLs. However, the measurement of small, dense LDLs requires sophisticated laboratory methods, such as ultracentrifugation, gradient gel electrophoresis, or nuclear magnetic resonance. We aimed to analyze whether the LDL apolipoprotein B (LDLapoB)-to-LDL cholesterol (LDLC) ratio is associated with cardiovascular mortality and whether this ratio represents a biomarker for small, dense LDLs. Methods: LDLC and LDLapoB were measured (beta-quantification) and calculated (according to Friedewald and Baca, respectively) for 3291 participants of the LURIC Study, with a median (inter-quartile range) follow-up for cardiovascular mortality of 9.9 (8.7−10.7) years. An independent replication cohort included 1660 participants. Associations of the LDLapoB/LDLC ratio with LDL subclass particle concentrations (ultracentrifugation) were tested for 282 participants. Results: In the LURIC Study, the mean (standard deviation) LDLC and LDLapoB concentrations were 117 (34) and 85 (22) mg/dL, respectively; 621 cardiovascular deaths occurred. Elevated LDLapoB/LDLC (calculated and measured) ratios were significantly and independently associated with increased cardiovascular mortality in the entire cohort (fourth vs. first quartile: hazard ratio (95% confidence interval) = 2.07 (1.53−2.79)) and in statin-naïve patients. The association between calculated LDLapoB/LDLC ratio and cardiovascular mortality was replicated in an independent cohort. High LDLapoB/LDLC ratios were associated with higher LDL5 and LDL6 concentrations (both p < 0.001), but not with concentrations of larger LDLs. Conclusions: Elevated measured and calculated LDLapoB/LDLC ratios are associated with increased cardiovascular mortality. Use of LDLapoB/LDLC ratios allows estimation of the atherogenic risk conferred by small, dense LDLs.
Collapse
|
34
|
Li Y, Cheng Y, Consolato F, Schiano G, Chong MR, Pietzner M, Nguyen NQH, Scherer N, Biggs ML, Kleber ME, Haug S, Göçmen B, Pigeyre M, Sekula P, Steinbrenner I, Schlosser P, Joseph CB, Brody JA, Grams ME, Hayward C, Schultheiss UT, Krämer BK, Kronenberg F, Peters A, Seissler J, Steubl D, Then C, Wuttke M, März W, Eckardt KU, Gieger C, Boerwinkle E, Psaty BM, Coresh J, Oefner PJ, Pare G, Langenberg C, Scherberich JE, Yu B, Akilesh S, Devuyst O, Rampoldi L, Köttgen A. Genome-wide studies reveal factors associated with circulating uromodulin and its relationships to complex diseases. JCI Insight 2022; 7:e157035. [PMID: 35446786 PMCID: PMC9220927 DOI: 10.1172/jci.insight.157035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/07/2022] [Indexed: 11/28/2022] Open
Abstract
Uromodulin (UMOD) is a major risk gene for monogenic and complex forms of kidney disease. The encoded kidney-specific protein uromodulin is highly abundant in urine and related to chronic kidney disease, hypertension, and pathogen defense. To gain insights into potential systemic roles, we performed genome-wide screens of circulating uromodulin using complementary antibody-based and aptamer-based assays. We detected 3 and 10 distinct significant loci, respectively. Integration of antibody-based results at the UMOD locus with functional genomics data (RNA-Seq, ATAC-Seq, Hi-C) of primary human kidney tissue highlighted an upstream variant with differential accessibility and transcription in uromodulin-synthesizing kidney cells as underlying the observed cis effect. Shared association patterns with complex traits, including chronic kidney disease and blood pressure, placed the PRKAG2 locus in the same pathway as UMOD. Experimental validation of the third antibody-based locus, B4GALNT2, showed that the p.Cys466Arg variant of the encoded N-acetylgalactosaminyltransferase had a loss-of-function effect leading to higher serum uromodulin levels. Aptamer-based results pointed to enzymes writing glycan marks present on uromodulin and to their receptors in the circulation, suggesting that this assay permits investigating uromodulin's complex glycosylation rather than its quantitative levels. Overall, our study provides insights into circulating uromodulin and its emerging functions.
Collapse
Affiliation(s)
- Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Francesco Consolato
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Michael R. Chong
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences and
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Maik Pietzner
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ngoc Quynh H. Nguyen
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nora Scherer
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Mary L. Biggs
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Marcus E. Kleber
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Burulça Göçmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Marie Pigeyre
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christina B. Joseph
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | | | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Nephrology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Jochen Seissler
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Dominik Steubl
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Cornelia Then
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner Munich, Neuherberg, Germany
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Epidemiology and
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, Washington, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Guillaume Pare
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Luca Rampoldi
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Moissl AP, Delgado GE, Krämer BK, Dawczynski C, Stojakovic T, März W, Kleber ME, Lorkowski S. Gender- and subgroup-specific sensitivity analysis of alcohol consumption and mortality in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. Data Brief 2022; 41:107873. [PMID: 35198663 PMCID: PMC8841993 DOI: 10.1016/j.dib.2022.107873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/19/2021] [Accepted: 01/21/2022] [Indexed: 10/26/2022] Open
|
36
|
Zeng L, Moser S, Mirza-Schreiber N, Lamina C, Coassin S, Nelson CP, Annilo T, Franzén O, Kleber ME, Mack S, Andlauer TFM, Jiang B, Stiller B, Li L, Willenborg C, Munz M, Kessler T, Kastrati A, Laugwitz KL, Erdmann J, Moebus S, Nöthen MM, Peters A, Strauch K, Müller-Nurasyid M, Gieger C, Meitinger T, Steinhagen-Thiessen E, März W, Metspalu A, Björkegren JLM, Samani NJ, Kronenberg F, Müller-Myhsok B, Schunkert H. Cis-epistasis at the LPA locus and risk of cardiovascular diseases. Cardiovasc Res 2022; 118:1088-1102. [PMID: 33878186 PMCID: PMC8930071 DOI: 10.1093/cvr/cvab136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 04/16/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Coronary artery disease (CAD) has a strong genetic predisposition. However, despite substantial discoveries made by genome-wide association studies (GWAS), a large proportion of heritability awaits identification. Non-additive genetic effects might be responsible for part of the unaccounted genetic variance. Here, we attempted a proof-of-concept study to identify non-additive genetic effects, namely epistatic interactions, associated with CAD. METHODS AND RESULTS We tested for epistatic interactions in 10 CAD case-control studies and UK Biobank with focus on 8068 SNPs at 56 loci with known associations with CAD risk. We identified a SNP pair located in cis at the LPA locus, rs1800769 and rs9458001, to be jointly associated with risk for CAD [odds ratio (OR) = 1.37, P = 1.07 × 10-11], peripheral arterial disease (OR = 1.22, P = 2.32 × 10-4), aortic stenosis (OR = 1.47, P = 6.95 × 10-7), hepatic lipoprotein(a) (Lp(a)) transcript levels (beta = 0.39, P = 1.41 × 10-8), and Lp(a) serum levels (beta = 0.58, P = 8.7 × 10-32), while individual SNPs displayed no association. Further exploration of the LPA locus revealed a strong dependency of these associations on a rare variant, rs140570886, that was previously associated with Lp(a) levels. We confirmed increased CAD risk for heterozygous (relative OR = 1.46, P = 9.97 × 10-32) and individuals homozygous for the minor allele (relative OR = 1.77, P = 0.09) of rs140570886. Using forward model selection, we also show that epistatic interactions between rs140570886, rs9458001, and rs1800769 modulate the effects of the rs140570886 risk allele. CONCLUSIONS These results demonstrate the feasibility of a large-scale knowledge-based epistasis scan and provide rare evidence of an epistatic interaction in a complex human disease. We were directed to a variant (rs140570886) influencing risk through additive genetic as well as epistatic effects. In summary, this study provides deeper insights into the genetic architecture of a locus important for cardiovascular diseases.
Collapse
Affiliation(s)
- Lingyao Zeng
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
| | - Sylvain Moser
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich 80804, Germany
| | - Nazanin Mirza-Schreiber
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Rd, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Tarmo Annilo
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Oscar Franzén
- Department of Genetics and Genomic Sciences and Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, 14186 Stockholm, Sweden
| | - Marcus E Kleber
- Medizinische Klinik V (Nephrologie, Hypertensiologie, Rheumatologie, Endokrinologie, Diabetologie), Medizinische Fakultät Mannheim der Universität Heidelberg, 69120 Heidelberg, Germany
| | - Salome Mack
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Till F M Andlauer
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Beibei Jiang
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Barbara Stiller
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
| | - Ling Li
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
| | - Christina Willenborg
- Institute for Cardiogenetics and University Heart Center Luebeck, University of Lübeck, 23562 Lübeck, Germany
| | - Matthias Munz
- Institute for Cardiogenetics and University Heart Center Luebeck, University of Lübeck, 23562 Lübeck, Germany
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
- Charité – University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Department of Periodontology and Synoptic Dentistry, 14197 Berlin, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics and University Heart Center Luebeck, University of Lübeck, 23562 Lübeck, Germany
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, 45147 Essen, Germany
- Centre for Urbane Epidemiology, University Hospital Essen, 45147 Essen, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, 53012 Bonn, Germany
| | - Annette Peters
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55101 Mainz, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55101 Mainz, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | | | - Winfried März
- Medizinische Klinik V (Nephrologie, Hypertensiologie, Rheumatologie, Endokrinologie, Diabetologie), Medizinische Fakultät Mannheim der Universität Heidelberg, 69120 Heidelberg, Germany
- Synlab Akademie, Synlab Holding Deutschland GmbH, Mannheim und Augsburg, 86156 Augsburg, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, 51010 Tartu, Estonia
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences and Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, 14186 Stockholm, Sweden
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Groby Rd, Leicester LE3 9QP, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Munich Cluster of Systems Biology, SyNergy, 81377 Munich, Germany
- Department of Health Data Science, University of Liverpool, Liverpool L69 3BX, UK
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, 80636 Munich, Germany
- Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK), Partner Site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| |
Collapse
|
37
|
Krohn JB, Nguyen YN, Akhavanpoor M, Erbel C, Domschke G, Linden F, Kleber ME, Delgado G, März W, Katus HA, Gleissner CA. Identification of Specific Coronary Artery Disease Phenotypes Implicating Differential Pathophysiologies. Front Cardiovasc Med 2022; 9:778206. [PMID: 35355960 PMCID: PMC8960070 DOI: 10.3389/fcvm.2022.778206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background and Aims The roles of multiple risk factors of coronary artery disease (CAD) are well established. Commonly, CAD is considered as a single disease entity. We wish to examine whether coronary angiography allows to identify distinct CAD phenotypes associated with major risk factors and differences in prognosis. Methods In a cohort of 4,344 patients undergoing coronary angiography at Heidelberg University Hospital between 2014 and 2016, cluster analysis of angiographic reports identified subgroups with similar patterns of spatial distribution of high-grade stenoses. Clusters were independently confirmed in 3,129 patients from the LURIC study. Results Four clusters were identified: cluster one lacking critical stenoses comprised the highest percentage of women with the lowest cardiovascular risk. Patients in cluster two exhibiting high-grade stenosis of the proximal RCA had a high prevalence of the metabolic syndrome, and showed the highest levels of inflammatory biomarkers. Cluster three with predominant proximal LAD stenosis frequently presented with acute coronary syndrome and elevated troponin levels. Cluster four with high-grade stenoses throughout had the oldest patients with the highest overall cardiovascular risk. All-cause and cardiovascular mortality differed significantly between the clusters. Conclusions We identified four phenotypic subgroups of CAD bearing distinct demographic and biochemical characteristics with differences in prognosis, which may indicate multiple disease entities currently summarized as CAD.
Collapse
Affiliation(s)
- Jona B. Krohn
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Y Nhi Nguyen
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Christian Erbel
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Gabriele Domschke
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Fabian Linden
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Marcus E. Kleber
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Graciela Delgado
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Winfried März
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Hugo A. Katus
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christian A. Gleissner
- Department of Cardiology, Pulmonology and Angiology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Cardiology and Stroke Centre, Rottal-Inn Kliniken, Eggenfelden, Germany
- *Correspondence: Christian A. Gleissner
| |
Collapse
|
38
|
Li L, Chen Z, von Scheidt M, Li S, Steiner A, Güldener U, Koplev S, Ma A, Hao K, Pan C, Lusis AJ, Pang S, Kessler T, Ermel R, Sukhavasi K, Ruusalepp A, Gagneur J, Erdmann J, Kovacic JC, Björkegren JLM, Schunkert H. Transcriptome-wide association study of coronary artery disease identifies novel susceptibility genes. Basic Res Cardiol 2022; 117:6. [PMID: 35175464 PMCID: PMC8852935 DOI: 10.1007/s00395-022-00917-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
The majority of risk loci identified by genome-wide association studies (GWAS) are in non-coding regions, hampering their functional interpretation. Instead, transcriptome-wide association studies (TWAS) identify gene-trait associations, which can be used to prioritize candidate genes in disease-relevant tissue(s). Here, we aimed to systematically identify susceptibility genes for coronary artery disease (CAD) by TWAS. We trained prediction models of nine CAD-relevant tissues using EpiXcan based on two genetics-of-gene-expression panels, the Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task (STARNET) and the Genotype-Tissue Expression (GTEx). Based on these prediction models, we imputed gene expression of respective tissues from individual-level genotype data on 37,997 CAD cases and 42,854 controls for the subsequent gene-trait association analysis. Transcriptome-wide significant association (i.e. P < 3.85e-6) was observed for 114 genes. Of these, 96 resided within previously identified GWAS risk loci and 18 were novel. Stepwise analyses were performed to study their plausibility, biological function, and pathogenicity in CAD, including analyses for colocalization, damaging mutations, pathway enrichment, phenome-wide associations with human data and expression-traits correlations using mouse data. Finally, CRISPR/Cas9-based gene knockdown of two newly identified TWAS genes, RGS19 and KPTN, in a human hepatocyte cell line resulted in reduced secretion of APOB100 and lipids in the cell culture medium. Our CAD TWAS work (i) prioritized candidate causal genes at known GWAS loci, (ii) identified 18 novel genes to be associated with CAD, and iii) suggested potential tissues and pathways of action for these TWAS CAD genes.
Collapse
Affiliation(s)
- Ling Li
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Fakultät für Informatik, Technische Universität München, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhifen Chen
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Moritz von Scheidt
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Shuangyue Li
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Andrea Steiner
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ulrich Güldener
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Angela Ma
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Calvin Pan
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shichao Pang
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Raili Ermel
- Department of Cardiac Surgery, The Heart Clinic, Tartu University Hospital, Tartu, Estonia
| | - Katyayani Sukhavasi
- Department of Cardiac Surgery, The Heart Clinic, Tartu University Hospital, Tartu, Estonia
| | - Arno Ruusalepp
- Department of Cardiac Surgery, The Heart Clinic, Tartu University Hospital, Tartu, Estonia
- Clinical Gene Networks AB, Stockholm, Sweden
| | - Julien Gagneur
- Fakultät für Informatik, Technische Universität München, Munich, Germany
| | - Jeanette Erdmann
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Jason C Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029-6574, USA
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
- Clinical Gene Networks AB, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany.
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
39
|
Frysz M, Gergei I, Scharnagl H, Smith GD, Zheng J, Lawlor DA, Herrmann M, Maerz W, Tobias JH. Circulating Sclerostin Levels Are Positively Related to Coronary Artery Disease Severity and Related Risk Factors. J Bone Miner Res 2022; 37:273-284. [PMID: 34738659 PMCID: PMC9377011 DOI: 10.1002/jbmr.4467] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Romosozumab is a newly available treatment for osteoporosis acting by sclerostin inhibition. Its cardiovascular safety has been questioned after finding excess cardiovascular disease (CVD)-related events in a pivotal phase 3 trial. Previous studies of relationships between circulating sclerostin levels and CVD and associated risk factors have yielded conflicting findings, likely reflecting small numbers and selected patient groups. We aimed to characterize relationships between sclerostin and CVD and related risk factors in more detail by examining these in two large cohorts, Ludwigshafen Risk and Cardiovascular Health study (LURIC; 34% female, mean age 63.0 years) and Avon Longitudinal Study of Parents and Children study (ALSPAC) mothers (mean age 48.1 years). Together these provided 5069 participants with complete data. Relationships between sclerostin and CVD risk factors were meta-analyzed, adjusted for age, sex (LURIC), body mass index, smoking, social deprivation, and ethnicity (ALSPAC). Higher sclerostin levels were associated with higher risk of diabetes mellitus (DM) (odds ratio [OR] = 1.25; 95% confidence interval [CI] 1.12, 1.37), risk of elevated fasting glucose (OR 1.15; CI 1.04, 1.26), and triglyceride levels (β 0.03; CI 0.00, 0.06). Conversely, higher sclerostin was associated with lower estimated glomerular filtration rate (eGFR) (β -0.20; CI -0.38, -0.02), HDL cholesterol (β -0.05; CI -0.10, -0.01), and apolipoprotein A-I (β -0.05; CI -0.08, -0.02) (difference in mean SD per SD increase in sclerostin, with 95% CI). In LURIC, higher sclerostin was associated with an increased risk of death from cardiac disease during follow-up (hazard ratio [HR] = 1.13; 1.03, 1.23) and with severity of coronary artery disease on angiogram as reflected by Friesinger score (0.05; 0.01, 0.09). Associations with cardiac mortality and coronary artery severity were partially attenuated after adjustment for risk factors potentially related to sclerostin, namely LDL and HDL cholesterol, log triglycerides, DM, hypertension, eGFR, and apolipoprotein A-I. Contrary to trial evidence suggesting sclerostin inhibition leads to an increased risk of CVD, sclerostin levels appear to be positively associated with coronary artery disease severity and mortality, partly explained by a relationship between higher sclerostin levels and major CVD risk factors. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Monika Frysz
- Musculoskeletal Research Unit, University of Bristol, Bristol, UK.,MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Ingrid Gergei
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Therapeutic Area Cardiovascular Medicine, Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Winfried Maerz
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - Jon H Tobias
- Musculoskeletal Research Unit, University of Bristol, Bristol, UK.,MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, UK
| |
Collapse
|
40
|
Genome-wide meta-analysis of phytosterols reveals five novel loci and a detrimental effect on coronary atherosclerosis. Nat Commun 2022; 13:143. [PMID: 35013273 PMCID: PMC8748632 DOI: 10.1038/s41467-021-27706-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022] Open
Abstract
Phytosterol serum concentrations are under tight genetic control. The relationship between phytosterols and coronary artery disease (CAD) is controversially discussed. We perform a genome-wide meta-analysis of 32 phytosterol traits reflecting resorption, cholesterol synthesis and esterification in six studies with up to 9758 subjects and detect ten independent genome-wide significant SNPs at seven genomic loci. We confirm previously established associations at ABCG5/8 and ABO and demonstrate an extended locus heterogeneity at ABCG5/8 with different functional mechanisms. New loci comprise HMGCR, NPC1L1, PNLIPRP2, SCARB1 and APOE. Based on these results, we perform Mendelian Randomization analyses (MR) revealing a risk-increasing causal relationship of sitosterol serum concentrations and CAD, which is partly mediated by cholesterol. Here we report that phytosterols are polygenic traits. MR add evidence of both, direct and indirect causal effects of sitosterol on CAD.
Collapse
|
41
|
Leiherer A, Mündlein A, Brandtner EM, Säly CH, Ramadani H, Vonbank A, Mader A, Dopheide JF, Jylhä A, Lääperi M, Laaksonen R, März W, Fraunberger P, Kleber M, Drexel H. Lipid profiles of patients with manifest coronary versus peripheral atherosclerosis - Is there a difference? J Intern Med 2021; 290:1249-1263. [PMID: 34337800 DOI: 10.1111/joim.13368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM Peripheral arterial disease (PAD) and coronary artery disease (CAD) are both caused by atherosclerosis. Serum lipids and lipoproteins are predictive of the development of atherosclerosis but it is not clear if they differ in the two manifestations, PAD and CAD. We tested whether a more detailed characterization of the lipid and lipoprotein patterns of PAD and CAD allows a clear differentiation between the two atherosclerotic phenotypes. METHODS A cohort of 274 statin-naïve patients with either newly diagnosed imaging proven PAD (n = 89) or stable CAD (n = 185) was characterized using nuclear magnetic resonance- and liquid chromatography-tandem mass spectrometry-based advanced lipid and lipoprotein analysis. An independent cohort of 1239 patients with PAD and CAD was used for validation. RESULTS We found a significant difference in markers of inflammation as well as ceramide and phosphatidylcholine levels between patients with PAD and CAD. In contrast, basic lipid markers including total cholesterol, LDL cholesterol, HDL cholesterol, lipoprotein(a) or detailed lipoprotein profiles did not differ significantly between patients with PAD and CAD. Applying ratios and scores derived from ceramides and phosphatidylcholines further improved the discrimination between PAD and CAD. These significant differences were independent of body composition, from the status of smoking or type 2 diabetes mellitus, and also from apolipoprotein C-III and other inflammatory parameters which were different between CAD and PAD. CONCLUSION The present study clearly suggests that PAD and CAD differ in terms of their ceramide- and phosphatidylcholine-based lipid patterns but not in lipoprotein characteristics.
Collapse
Affiliation(s)
- Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein.,Medical Central Laboratories, Feldkirch, Austria
| | - Axel Mündlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Eva Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Christoph H Säly
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Department of Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Hana Ramadani
- Division of Angiology, Swiss Cardiovascular Center, University Hospital of Bern, Bern, Switzerland
| | - Alexander Vonbank
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Department of Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Arthur Mader
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Department of Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Jörn F Dopheide
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Division of Angiology, Swiss Cardiovascular Center, University Hospital of Bern, Bern, Switzerland
| | | | | | - Reijo Laaksonen
- Zora Biosciences, Espoo, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, Finland
| | - Winfried März
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Holding, Mannheim, Germany
| | - Peter Fraunberger
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein.,Medical Central Laboratories, Feldkirch, Austria
| | - Marcus Kleber
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.,Division of Angiology, Swiss Cardiovascular Center, University Hospital of Bern, Bern, Switzerland.,Private University in the Principality of Liechtenstein, Triesen, Liechtenstein.,Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Department of Internal Medicine, Academic Teaching Hospital Bregenz, Bregenz, Austria
| |
Collapse
|
42
|
Laaksonen J, Mishra PP, Seppälä I, Raitoharju E, Marttila S, Mononen N, Lyytikäinen LP, Kleber ME, Delgado GE, Lepistö M, Almusa H, Ellonen P, Lorkowski S, März W, Hutri-Kähönen N, Raitakari O, Kähönen M, Salonen JT, Lehtimäki T. Mitochondrial genome-wide analysis of nuclear DNA methylation quantitative trait loci. Hum Mol Genet 2021; 31:1720-1732. [PMID: 35077545 PMCID: PMC9122653 DOI: 10.1093/hmg/ddab339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Abstract
Mitochondria have a complex communication network with the surrounding cell and can alter nuclear DNA methylation (DNAm). Variation in the mitochondrial DNA (mtDNA) has also been linked to differential DNAm. Genome-wide association studies have identified numerous DNAm quantitative trait loci, but these studies have not examined the mitochondrial genome. Herein, we quantified nuclear DNAm from blood and conducted a mitochondrial genome-wide association study of DNAm, with an additional emphasis on sex- and prediabetes-specific heterogeneity. We used the Young Finns Study (n = 926) with sequenced mtDNA genotypes as a discovery sample and sought replication in the Ludwigshafen Risk and Cardiovascular Health study (n = 2317). We identified numerous significant associations in the discovery phase (P < 10−9), but they were not replicated when accounting for multiple testing. In total, 27 associations were nominally replicated with a P < 0.05. The replication analysis presented no evidence of sex- or prediabetes-specific heterogeneity. The 27 associations were included in a joint meta-analysis of the two cohorts, and 19 DNAm sites associated with mtDNA variants, while four other sites showed haplogroup associations. An expression quantitative trait methylation analysis was performed for the identified DNAm sites, pinpointing two statistically significant associations. This study provides evidence of a mitochondrial genetic control of nuclear DNAm with little evidence found for sex- and prediabetes-specific effects. The lack of a comparable mtDNA data set for replication is a limitation in our study and further studies are needed to validate our results.
Collapse
Affiliation(s)
- Jaakko Laaksonen
- To whom correspondence should be addressed at: 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, Tampere FI-33014, Finland. Tel: +358 504080774; E-mail:
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Saara Marttila
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
- Gerontology Research Center, Tampere University, Tampere 33520, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Maija Lepistö
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki 00290, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki 00290, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki 00290, Finland
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena 07743, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena 07743, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena 07743, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg 86156, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8010, Austria
| | - Nina Hutri-Kähönen
- Tampere Centre for Skills Training and Simulation, Tampere University, Tampere 33520, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku 20520, Finland
- Research Centre for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33520, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Jukka T Salonen
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- MAS-Metabolic Analytical Services Oy, Helsinki 00990, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| |
Collapse
|
43
|
Hall IF, Climent M, Viviani Anselmi C, Papa L, Tragante V, Lambroia L, Farina FM, Kleber ME, März W, Biguori C, Condorelli G, Elia L. rs41291957 controls miR-143 and miR-145 expression and impacts coronary artery disease risk. EMBO Mol Med 2021; 13:e14060. [PMID: 34551209 PMCID: PMC8495461 DOI: 10.15252/emmm.202114060] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 01/25/2023] Open
Abstract
The role of single nucleotide polymorphisms (SNPs) in the etiopathogenesis of cardiovascular diseases is well known. The effect of SNPs on disease predisposition has been established not only for protein coding genes but also for genes encoding microRNAs (miRNAs). The miR-143/145 cluster is smooth muscle cell-specific and implicated in the pathogenesis of atherosclerosis. Whether SNPs within the genomic sequence of the miR-143/145 cluster are involved in cardiovascular disease development is not known. We thus searched annotated sequence databases for possible SNPs associated with miR-143/145. We identified one SNP, rs41291957 (G > A), located -91 bp from the mature miR-143 sequence, as the nearest genetic variation to this miRNA cluster, with a minor allele frequency > 10%. In silico and in vitro approaches determined that rs41291957 (A) upregulates miR-143 and miR-145, modulating phenotypic switching of vascular smooth cells towards a differentiated/contractile phenotype. Finally, we analysed association between rs41291957 and CAD in two cohorts of patients, finding that the SNP was a protective factor. In conclusion, our study links a genetic variation to a pathological outcome through involvement of miRNAs.
Collapse
Affiliation(s)
- Ignacio Fernando Hall
- Humanitas Research Hospital‐IRCCSRozzanoItaly
- Institute of Genetics and Biomedical ResearchNational Research CouncilRozzanoItaly
| | | | | | - Laura Papa
- Humanitas Research Hospital‐IRCCSRozzanoItaly
| | - Vinicius Tragante
- Department of CardiologyDivision Heart and LungsUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Luca Lambroia
- Humanitas Research Hospital‐IRCCSRozzanoItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Floriana Maria Farina
- Humanitas Research Hospital‐IRCCSRozzanoItaly
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximillians‐Universität (LMU) MünchenMunichGermany
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Marcus E Kleber
- V Department of MedicineMedical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Winfried März
- V Department of MedicineMedical Faculty MannheimHeidelberg UniversityMannheimGermany
- SYNLAB AcademySYNLAB Holding Deutschland GmbHAugsburg and MannheimGermany
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University GrazGrazAustria
| | - Carlo Biguori
- Interventional Cardiology UnitMediterranea CardiocentroNaplesItaly
| | - Gianluigi Condorelli
- Humanitas Research Hospital‐IRCCSRozzanoItaly
- Institute of Genetics and Biomedical ResearchNational Research CouncilRozzanoItaly
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
| | - Leonardo Elia
- Humanitas Research Hospital‐IRCCSRozzanoItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| |
Collapse
|
44
|
Pott J, Gadin J, Theusch E, Kleber ME, Delgado GE, Kirsten H, Hauck SM, Burkhardt R, Scharnagl H, Krauss RM, Loeffler M, März W, Thiery J, Silveira A, Vant Hooft FM, Scholz M. Meta-GWAS of PCSK9 levels detects two novel loci at APOB and TM6SF2. Hum Mol Genet 2021; 31:999-1011. [PMID: 34590679 PMCID: PMC8947322 DOI: 10.1093/hmg/ddab279] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/15/2022] Open
Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key player in lipid metabolism, as it degrades low-density lipoprotein (LDL) receptors from hepatic cell membranes. So far, only variants of the PCSK9 gene locus were found to be associated with PCSK9 levels. Here we aimed to identify novel genetic loci that regulate PCSK9 levels and how they relate to other lipid traits. Additionally, we investigated to what extend the causal effect of PCSK9 on coronary artery disease (CAD) is mediated by low-density lipoprotein–cholesterol (LDL–C). Methods and Results We performed a genome-wide association study meta-analysis of PCSK9 levels in up to 12 721 samples of European ancestry. The estimated heritability was 10.3%, which increased to 12.6% using only samples from patients without statin treatment. We successfully replicated the known PCSK9 hit consisting of three independent signals. Interestingly, in a study of 300 African Americans, we confirmed the locus with a different PCSK9 variant. Beyond PCSK9, our meta-analysis detected three novel loci with genome-wide significance. Co-localization analysis with cis-eQTLs and lipid traits revealed biologically plausible candidate genes at two of them: APOB and TM6SF2. In a bivariate Mendelian Randomization analysis, we detected a strong effect of PCSK9 on LDL-C, but not vice versa. LDL-C mediated 63% of the total causal effect of PCSK9 on CAD. Conclusion Our study identified novel genetic loci with plausible candidate genes affecting PCSK9 levels. Ethnic heterogeneity was observed at the PCSK9 locus itself. Although the causal effect of PCSK9 on CAD is mainly mediated by LDL-C, an independent direct effect also occurs.
Collapse
Affiliation(s)
- Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany.,LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Jesper Gadin
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital Solna, Sweden
| | - Elizabeth Theusch
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany.,LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core and Research Unit Protein Science, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ralph Burkhardt
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig.,Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Ronald M Krauss
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA.,Department of Medicine, University of California San Francisco, Oakland, CA, USA
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany.,LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,SYNLAB Academy, SYNALB Holding Deutschland GmbH, Mannheim, Germany
| | - Joachim Thiery
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig.,Faculty of Medicine, Kiel University, Kiel, Germany
| | - Angela Silveira
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital Solna, Sweden
| | - Ferdinand M Vant Hooft
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Karolinska University Hospital Solna, Sweden
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany.,LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
45
|
Yazdani B, Delgado GE, Scharnagl H, Krämer BK, Drexel H, März W, Scherberich JE, Leiherer A, Kleber ME. Combined Use of Serum Uromodulin and eGFR to Estimate Mortality Risk. Front Med (Lausanne) 2021; 8:723546. [PMID: 34568379 PMCID: PMC8455921 DOI: 10.3389/fmed.2021.723546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Serum uromodulin (sUmod) shows a strong direct correlation with eGFR in patients with impaired kidney function and an inverse association with mortality. However, there are patients in whom only one of both markers is decreased. Therefore, we aimed to investigate the effect of marker discordance on mortality risk. sUmod and eGFR were available in 3,057 participants of the Ludwigshafen Risk and Cardiovascular Health study and 529 participants of the VIVIT study. Both studies are monocentric prospective studies of patients that had been referred for coronary angiography. Participants were categorized into four groups according to the median values of sUmod (LURIC: 146 ng/ml, VIVIT: 156) and eGFR (LURIC: 84 ml/min/1.73 m2, VIVIT: 87). In 945 LURIC participants both markers were high (UHGH), in 935 both were low (ULGL), in 589 only eGFR (UHGL), and in 582 only sUmod (ULGH) was low. After balancing the groups for cardiovascular risk factors, hazard ratios (95%CI) for all-cause mortality as compared to UHGH were 2.03 (1.63-2.52), 1.43 (1.13-1.81), and 1.32 (1.03-1.69) for ULGL, UHGL, and ULGH, respectively. In VIVIT, HRs were 3.12 (1.38-7.08), 2.38 (1.01-5.61), and 2.06 (0.81-5.22). Adding uromodulin to risk prediction models that already included eGFR as a covariate slightly increased the Harrell's C and significantly improved the AUC in LURIC. In UHGL patients, hypertension, heart failure and upregulation of the renin-angiotensin-aldosterone-system seem to be the driving forces of disease development, whereas in ULGH patients metabolic disturbances might be key drivers of increased mortality. In conclusion, SUmod/eGFR subgroups mirror distinct metabolic and clinical patterns. Assessing sUmod additionally to creatinine or cystatin C has the potential to allow a more precise risk modeling and might improve risk stratification.
Collapse
Affiliation(s)
- Babak Yazdani
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Bernhard K Krämer
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience, Mannheim, Germany
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment at the Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Liechtenstein.,Drexel University College of Medicine, Philadelphia, PA, United States.,Division of Angiology, Swiss Cardiovascular Center, University Hospital of Bern, Bern, Switzerland
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Jürgen E Scherberich
- Klinikum München-Harlaching, Teaching Hospital of the Ludwig-Maximilians University, Munich, Germany.,KfH-München Süd, Munich, Germany
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment at the Academic Teaching Hospital Feldkirch, Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Liechtenstein.,Medical Central Laboratories, Feldkirch, Austria
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| |
Collapse
|
46
|
Gergei I, Zheng J, Andlauer TFM, Brandenburg V, Mirza-Schreiber N, Müller-Myhsok B, Krämer BK, Richard D, Falk L, Movérare-Skrtic S, Ohlsson C, Smith GD, März W, Voelkl J, Tobias JH. GWAS META-analysis followed by MENDELIAN randomisation revealed potential control mechanisms for circulating α-klotho levels. Hum Mol Genet 2021; 31:792-802. [PMID: 34542150 PMCID: PMC8895756 DOI: 10.1093/hmg/ddab263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/08/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The protein α-Klotho acts as transmembrane the co-receptor for fibroblast growth factor 23 (FGF-23) and is a key regulator of phosphate homeostasis. However, α-Klotho also exists in a circulating form, with pleiotropic, but incompletely understood functions and regulation. Therefore, we undertook a GWAS meta-analysis followed by Mendelian randomisation (MR) of circulating α-Klotho levels. METHODS Plasma α-Klotho levels were measured by ELISA in the LURIC and ALSPAC (mothers) cohorts, followed by a GWAS meta-analysis in 4376 individuals across the two cohorts. RESULTS Six signals at five loci were associated with circulating α-Klotho levels at genome-wide significance (p < 5 × 10-8), namely ABO, KL, FGFR1, and two post-translational modification genes, B4GALNT3 and CHST9. Together, these loci explained > 9% of the variation in circulating α-Klotho levels. MR analyses revealed no causal relationships between α-Klotho and renal function, FGF-23-dependent factors such as vitamin D and phosphate levels, or bone mineral density. The screening for genetic correlations with other phenotypes, followed by targeted MR suggested causal effects of liability of Crohn's disease risk [IVW beta = 0.059 (95% CI 0.026, 0.093)] and low-density lipoprotein cholesterol (LDL-C) levels [-0.198, (-0.332, -0.063)] on α-Klotho. CONCLUSIONS Our GWAS findings suggest that two enzymes involved in post-translational modification, B4GALNT3 and CHST9, contribute to genetic influences on α-Klotho levels, presumably by affecting protein turnover and stability. Subsequent evidence from MR analyses on α-Klotho levels suggest regulation by mechanisms besides phosphate-homeostasis and raise the possibility of cross-talk with FGF19- and FGF21-dependent pathways, respectively.
Collapse
Affiliation(s)
- Ingrid Gergei
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Therapeutic Area Cardiovascular Medicine, Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | | | | | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bernhard K Krämer
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,European Center for Angioscience ECAS, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, USA
| | - Louise Falk
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
| | - Sofia Movérare-Skrtic
- University of Gothenburg, Sahlgrenska Osteoporosis Centre, CBAR, Institute of Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden
| | - Claes Ohlsson
- University of Gothenburg, Sahlgrenska Osteoporosis Centre, CBAR, Institute of Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Drug Treatment, Gothenburg, Sweden
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria.,Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Musculoskeletal Research Unit, Translational HeaalthLevel 1 Learning and Research Building, Southmead Hospital, Bristol, United Kingdom
| |
Collapse
|
47
|
PAQR6 Upregulation Is Associated with AR Signaling and Unfavorite Prognosis in Prostate Cancers. Biomolecules 2021; 11:biom11091383. [PMID: 34572596 PMCID: PMC8465620 DOI: 10.3390/biom11091383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
Progesterone-induced rapid non-genomic signaling events have been confirmed through several membrane progesterone receptors (mPR). Some mPRs were reported to correlate with cancer progression and patient prognosis. In this study, we conducted a comprehensive analysis of all progesterone receptor (PGR)-related genes in prostate cancer tissues and examined the correlations of their expression levels with disease progression and patient survival outcomes. We utilized multiple RNA-seq and cDNA microarray datasets to analyze gene expression profiles and performed logistics aggression and Kaplan-Meier survival analysis after stratifying patients based on tumor stages and Gleason scores. We also used NCBI GEO datasets to examine gene expression patterns in individual cell types of the prostate gland and to determine the androgen-induced alteration of gene expression. Spearman coefficient analysis was conducted to access the correlation of target gene expression with treatment responses and disease progression status. The classic PGR was mainly expressed in stromal cells and progestin and adipoQ receptor (PAQR) genes were the predominant genes in prostate epithelial cells. Progesterone receptor membrane component-1 (PGRMC1) was significantly higher than PGRMC2 in all prostate cell types. In prostate cancer tissues, PAQR6 expression was significantly upregulated, while all other genes were largely downregulated compared to normal prostate tissues. Although both PAQR6 upregulation and PAQR5 downregulation were significantly correlated with tumor pathological stages, only PAQR6 upregulation was associated with Gleason score, free-prostate-specific antigen (fPSA)/total-PSA (tPSA) ratio, and patient overall survival outcomes. In addition, PAQR6 upregulation and PGR/PGRMC1 downregulation were significantly associated with a quick relapse. Conversely, in neuroendocrinal prostate cancer (NEPC) tissues, PAQR6 expression was significantly lower, but PAQR7/8 expression was higher than castration-resistant prostate cancer (CRPC) tissues. PAQR8 expression was positively correlated with androgen receptor (AR) score and AR-V7 expression levels but inversely correlated with NEPC score in metastatic CRPC tumors. This study provides detailed expression profiles of membrane progesterone receptor genes in primary cancer, CRPC, and NEPC tissues. PAQR6 upregulation in primary cancer tissues is a novel prognostic biomarker for disease progression, overall, and progression-free survival in prostate cancers. PAQR8 expression in CRPC tissues is a biomarker for AR activation.
Collapse
|
48
|
Delgado GE, Kleber ME, Moissl AP, Yazdani B, Kusnik A, Ebert MP, März W, Krämer BK, Lammert A, Teufel A. Surrogate scores of advanced fibrosis in NAFLD/NASH do not predict mortality in patients with medium-to-high cardiovascular risk. Am J Physiol Gastrointest Liver Physiol 2021; 321:G252-G261. [PMID: 34132110 DOI: 10.1152/ajpgi.00058.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Untreated non-alcoholic fatty liver disease (NAFLD) may have significant consequences including an increase in mortality and cardiovascular injury. Thus, early detection of NAFLD is currently believed not only to prevent liver-related but also cardiovascular mortality. However, almost nothing is known about coexisting NAFLD in patients with coronary artery disease (CAD). We investigated the impact of surrogate scores of fibrosis in NAFLD in a large cohort of patients referred to coronary angiography. Modeling the common NALFD and fibrosis scores, fibrosis-4 index (FIB-4) and NAFLD fibrosis score (NFS), as splines revealed significant associations with all-cause and cardiovascular mortality when Cox regression models were only adjusted for cardiovascular risk factors that were not already included in the calculation of the scores. Stratifying the scores into quartiles yielded hazard ratios [95% confidence interval (CI)] for all-cause and cardiovascular mortality for the 4th quartile versus the 1st quartile of 2.28 (1.90-2.75) and 2.11 (1.67-2.67) for FIB-4 and of 3.21 (2.61-3.94) and 3.12 (2.41-4.04) for NFS. However, we did not observe an independent association of FIB-4 or NFS with overall or cardiovascular mortality in our prospective CAD cohort after full adjustment for all cardiovascular risk factors [all-cause mortality: HR 1.13 (0.904-1.41) and 1.17 (0.903-1.52); cardiovascular mortality: HR 1.06 (0.8-1.41) and 1.02 (0.738-1.41)]. Thus, neither FIB-4 nor NFS, as surrogate markers for NAFLD/NASH, were independent risk factors for overall or cardiovascular mortality in patients with CAD. Our data show that surrogate risk scores for NAFLD-related fibrosis do not add information in assessing the CVD events in patients with CAD proven by angiography.NEW & NOTEWORTHY We investigated the impact of NAFLD surrogate markers in a large cohort of patients that had been referred to coronary angiography. In contrast to a repeatedly demonstrated increased link of cardiovascular events in patients with NALFD, we demonstrated that NAFLD surrogate markers were not independent risk factors for overall or cardiovascular mortality in patients with CAD. Thus, these markers may not be useful for primary prevention of cardiovascular events in patients with CAD.
Collapse
Affiliation(s)
- Graciela E Delgado
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marcus E Kleber
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Angela P Moissl
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany.,Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany
| | - Babak Yazdani
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Kusnik
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Winfried März
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Bernhard K Krämer
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Lammert
- Department of Medicine V, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Praxis für Stoffwechsel- und Nierenerkrankungen, Zentrum für Dialyse und Apherese, Grünstadt, Germany
| | - Andreas Teufel
- Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Medicine II, Section of Hepatology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
49
|
Lanser L, Fuchs D, Scharnagl H, Grammer T, Kleber ME, März W, Weiss G, Kurz K. Anemia of Chronic Disease in Patients With Cardiovascular Disease. Front Cardiovasc Med 2021; 8:666638. [PMID: 34458328 PMCID: PMC8387678 DOI: 10.3389/fcvm.2021.666638] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/23/2021] [Indexed: 12/28/2022] Open
Abstract
Objective: Anemia is often found in patients with coronary artery disease (CAD) or acute coronary syndrome (ACS) and related to disease severity. Our study investigated the relationship between anemia, iron homeostasis and inflammation in CAD and examined their influence on the outcome of patients. Patients and Methods: Markers of immune activation (neopterin, interleukin [IL]-12, IL-6, high sensitive C-reactive protein (hsCRP), fibrinogen, serum amyloid A [SAA]) and iron metabolism (ferritin, transferrin saturation, hemoglobin) were determined in 2,082 patients (68.7 % men, median age 63 years) from the Ludwigshafen Risk and cardiovascular Health (LURIC) cohort. Patients were followed-up for a median of 9.81 years. Results: 960 patients (46.1 %) presented with chronic CAD, 645 patients (31.0 %) had an ACS, and 477 patients (22.9 %) presented with no CAD in coronary angiography (CAG). Anemia (n = 357, 17.1 %) was associated with disease severity (reflected by more progressed stenosis in CAG, CCS, and NYHA classes, and a lower LV-EF), a higher cardio-cerebrovascular event rate and higher levels of inflammatory markers. Interestingly, anemia was only predictive for an adverse outcome in patients with elevated inflammatory markers. Accordingly, anemia of chronic disease (ACD) was associated with a higher cardio-cerebrovascular event-rate in the subsequent 2 years as compared to patients with other types of anemia or without anemia (14.3 vs. 6.1 vs. 4.0%, p < 0.001). Conclusions: This study confirms that anemia and immune activation are strongly related to cardiovascular disease progression and an adverse outcome. Our data suggest that the association of anemia with disease severity and outcome might mainly be due to underlying inflammation.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Graz Medical University, Graz, Austria
| | - Tanja Grammer
- Medical CIinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcus E Kleber
- Medical CIinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Winfried März
- Medical CIinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Günter Weiss
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
50
|
Moissl AP, Delgado GE, Krämer BK, Dawczynski C, Stojakovic T, März W, Kleber ME, Lorkowski S. Alcohol consumption and mortality: The Ludwigshafen Risk and Cardiovascular Health (LURIC) study. Atherosclerosis 2021; 335:119-125. [PMID: 34454737 DOI: 10.1016/j.atherosclerosis.2021.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/04/2021] [Accepted: 08/10/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS One of the most important risk factors for morbidity and mortality is the consumption of alcohol. The aim of our study was to examine the effect of alcohol consumption on all-cause mortality and cardiovascular mortality. METHODS The Ludwigshafen Risk and Cardiovascular Health (LURIC) study includes 3316 patients hospitalized for coronary angiography at a tertiary care centre in Southwest Germany. Patients were followed-up for a median of 9.9 (range 0.1-11.9 years) years. Total mortality number in the follow-up period was 995, and the number of incident cases, i.e. cardiovascular death, was 622. Information on alcohol consumption assessed by self-report questionnaires was used to calculate intake in grams of ethanol per day. Associations of alcohol consumption with morbidity and mortality were analysed using Cox proportional hazards regression. RESULTS We found significantly increased mortality for patients in the highest alcohol intake group age- and sex-adjusted (hazard ratio of 1.59 (95%CI, 0.93-2.72)) and a reduced risk for the group of low-volume drinkers (hazard ratio of 0.75 (95%CI, 0.65-0.86)). After adjustment for cardiovascular risk factors, the risk difference between abstainers and low-volume drinkers was not significant anymore. CONCLUSIONS In the LURIC study, the risk of overall mortality and cardiovascular mortality is significantly increased in study participants with very high alcohol consumption and slightly increased in total abstainers as compared to participants with low consumption in unadjusted analysis, replicating the well-known J-curve. Adjusting for cardiovascular risk factors rendered the risk decrease observed for low-volume drinkers insignificant. Therefore, our results do not show a significant health benefit of low-volume alcohol consumption in a cohort of patients at medium-to-high cardiovascular risk.
Collapse
Affiliation(s)
- Angela P Moissl
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany; Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany.
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bernhard K Krämer
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christine Dawczynski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Graz, Austria
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany.
| |
Collapse
|