101
|
Huet F, Dupuy AM, Duflos C, Reis CA, Kuster N, Aguilhon S, Cristol JP, Roubille F. Soluble urokinase-type plasminogen activator receptor strongly predicts global mortality in acute heart failure patients: insight from the STADE-HF registry. Future Sci OA 2021; 7:FSO697. [PMID: 34046195 PMCID: PMC8147742 DOI: 10.2144/fsoa-2020-0197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Whether soluble urokinase-type plasminogen activator receptor (suPAR) could be a valuable prognostic indicator remains uncertain. Materials & methods Patients from STADE-HF (Soluble Suppression of Tumorigenesis-2 as a Help for Management of Diagnosis, Evaluation and Management of Heart Failure) were included for analysis. Results 95 patients were included. The suPAR level of expression was significantly higher in the group of patients who died at one month (7.90 ± 4.35 ng/ml vs 11.94 ± 6.86 ng/ml; p < 0.05) or 1 year (7.28 ± 4.27 ng/ml vs 11.81 ± 4.88 ng/ml; p < 0.01), but there was no significant difference according to the readmission. Conclusion High suPAR levels during hospitalization for acute heart failure were highly predictive for the risk of mortality, but not the risk of readmission.
Collapse
Affiliation(s)
- Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| | - Anne-Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, 34090, France
| | - Claire Duflos
- Medico-Economic Research Unit, CHU Montpellier, University of Montpellier, Montpellier, 34090, France
| | - Cintia Azara Reis
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Nils Kuster
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France.,Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, 34090, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| | - François Roubille
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214, Montpellier, 34090, France
| |
Collapse
|
102
|
Lau ES, Paniagua SM, Liu E, Jovani M, Li SX, Takvorian K, Suthahar N, Cheng S, Splansky GL, Januzzi JL, Wang TJ, Vasan RS, Kreger B, Larson MG, Levy D, de Boer RA, Ho JE. Cardiovascular Risk Factors are Associated with Future Cancer. JACC: CARDIOONCOLOGY 2021; 3:48-58. [PMID: 33870217 PMCID: PMC8045786 DOI: 10.1016/j.jaccao.2020.12.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The extent to which co-occurrence of cardiovascular disease (CVD) and cancer is due to shared risk factors or other mechanisms is unknown. Objectives This study investigated the association of standard CVD risk factors, CVD biomarkers, pre-existing CVD, and ideal cardiovascular (CV) health metrics with the development of future cancer. Methods This study prospectively followed Framingham Heart Study and PREVEND (Prevention of Renal and Vascular End-Stage Disease) study participants free of cancer at baseline and ascertained histology-proven cancer. This study assessed the association of baseline CV risk factors, 10-year atherosclerotic (ASCVD) risk score, established CVD biomarkers, prevalent CVD, and the American Heart Association (AHA) Life’s Simple 7 CV health score with incident cancer using multivariable Cox models. Analyses of interim CVD events with incident cancer used time-dependent covariates. Results Among 20,305 participants (mean age 50 ± 14 years; 54% women), 2,548 incident cancer cases occurred over a median follow-up of 15.0 years (quartile 1 to 3: 13.3 to 15.0 years). Traditional CVD risk factors, including age, sex, and smoking status, were independently associated with cancer (p < 0.001 for all). Estimated 10-year ASCVD risk was also associated with future cancer (hazard ratio [HR]: 1.16 per 5% increase in risk; 95% confidence interval [CI] 1.14 to 1.17; p < 0.001). The study found that natriuretic peptides (tertile 3 vs. tertile 1; HR: 1.40; 95% CI: 1.03 to 1.91; p = 0.035) were associated with incident cancer but not high-sensitivity troponin (p = 0.47). Prevalent CVD and the development of interim CV events were not associated with higher risk of subsequent cancer. However, ideal CV health was associated with lower future cancer risk (HR: 0.95 per 1-point increase in the AHA health score; 95% CI: 0.92 to 0.99; p = 0.009). Conclusions CVD risk, as captured by traditional CVD risk factors, 10-year ASCVD risk score, and natriuretic peptide concentrations are associated with increased risk of future cancer. Conversely, a heart healthy lifestyle is associated with a lower risk of future cancer. These data suggest that the association between CVD and future cancer is attributable to shared risk factors.
Collapse
Affiliation(s)
- Emily S. Lau
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Samantha M. Paniagua
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elizabeth Liu
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Manol Jovani
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Shawn X. Li
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katherine Takvorian
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Navin Suthahar
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susan Cheng
- Department of Cardiology, Cedars Sinai Medical Center, Los Angeles, California, USA
| | | | - James L. Januzzi
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas J. Wang
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ramachandran S. Vasan
- Framingham Heart Study, Framingham, Massachusetts, USA
- Cardiovascular Medicine Section, Department of Medicine ad Section of Preventive Medicine and Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Bernard Kreger
- Cardiovascular Medicine Section, Department of Medicine ad Section of Preventive Medicine and Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Martin G. Larson
- Framingham Heart Study, Framingham, Massachusetts, USA
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Daniel Levy
- Framingham Heart Study, Framingham, Massachusetts, USA
- Center for Population Studies of the National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Rudolf A. de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jennifer E. Ho
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Address for correspondence: Dr. Jennifer E. Ho, Massachusetts General Hospital, 185 Cambridge Street, CPZN #3192, Boston, Massachusetts 02114, USA. @JenHoCardiology@JJheartdoc
| |
Collapse
|
103
|
Kim AJ, Ro H, Kim H, Chang JH, Lee HH, Chung W, Jung JY. Soluble ST2 and Galectin-3 as Predictors of Chronic Kidney Disease Progression and Outcomes. Am J Nephrol 2021; 52:119-130. [PMID: 33725696 DOI: 10.1159/000513663] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/04/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Soluble suppression of tumorigenicity-2 (sST2) and galectin-3, novel biomarkers of heart failure and cardiovascular stress, predict cardiovascular events (CVEs) and mortality. However, their relationship with kidney function and adverse outcomes in CKD are uncertain. The purpose of this study was to determine the association between sST2 and galectin-3 with CKD progression and adverse clinical outcomes. METHODS We measured baseline sST2 and galectin-3 levels in the CKD patient cohort at our institution between October 2013 and December 2014. The primary outcome was CKD progression (kidney failure with replacement therapy or ≥50% reduction in estimated glomerular filtration rate from the baseline). The secondary outcome was the composite of CVEs and death. We used a Cox proportional hazards model to evaluate the associations between sST2 and galectin-3 levels, with kidney and clinical outcomes. RESULTS In total, 352 patients were enrolled in this study. At baseline, log sST2 and galectin-3 were directly associated with the serum creatinine (Cr) and urine protein-to-Cr ratio. Cox regression analysis showed that the baseline log sST2 level independently predicted CKD progression and composite outcome after adjustment for age, sex, smoking, diabetes mellitus, hypertension, cardiovascular disease, renin-angiotensin system blocker, calcium channel blocker, β-blocker, diuretics, antiplatelet agents, anemia, and hypoalbuminemia. The baseline log galectin-3 level was independently associated with CKD progression, but not with the composite outcome after adjustment for confounding variables. CONCLUSIONS Elevated levels of sST2 and galectin-3 are significantly associated with CKD progression, but only sST2 is associated with adverse clinical outcomes.
Collapse
Affiliation(s)
- Ae Jin Kim
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Han Ro
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Hyunsook Kim
- Department of Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Jae Hyun Chang
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Hyun Hee Lee
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Wookyung Chung
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Ji Yong Jung
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea,
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea,
- Department of Health Sciences and Technology, Gachon University, Incheon, Republic of Korea,
| |
Collapse
|
104
|
Stege NM, de Boer RA, van den Berg MP, Silljé HHW. The Time Has Come to Explore Plasma Biomarkers in Genetic Cardiomyopathies. Int J Mol Sci 2021; 22:2955. [PMID: 33799487 PMCID: PMC7998409 DOI: 10.3390/ijms22062955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
For patients with hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) or arrhythmogenic cardiomyopathy (ACM), screening for pathogenic variants has become standard clinical practice. Genetic cascade screening also allows the identification of relatives that carry the same mutation as the proband, but disease onset and severity in mutation carriers often remains uncertain. Early detection of disease onset may allow timely treatment before irreversible changes are present. Although plasma biomarkers may aid in the prediction of disease onset, monitoring relies predominantly on identifying early clinical symptoms, on imaging techniques like echocardiography (Echo) and cardiac magnetic resonance imaging (CMR), and on (ambulatory) electrocardiography (electrocardiograms (ECGs)). In contrast to most other cardiac diseases, which are explained by a combination of risk factors and comorbidities, genetic cardiomyopathies have a clear primary genetically defined cardiac background. Cardiomyopathy cohorts could therefore have excellent value in biomarker studies and in distinguishing biomarkers related to the primary cardiac disease from those related to extracardiac, secondary organ dysfunction. Despite this advantage, biomarker investigations in cardiomyopathies are still limited, most likely due to the limited number of carriers in the past. Here, we discuss not only the potential use of established plasma biomarkers, including natriuretic peptides and troponins, but also the use of novel biomarkers, such as cardiac autoantibodies in genetic cardiomyopathy, and discuss how we can gauge biomarker studies in cardiomyopathy cohorts for heart failure at large.
Collapse
Affiliation(s)
| | | | | | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, AB43, 9713 GZ Groningen, The Netherlands; (N.M.S.); (R.A.d.B.); (M.P.v.d.B.)
| |
Collapse
|
105
|
Salzano A, D'Assante R, Israr MZ, Eltayeb M, D'Agostino A, Bernieh D, De Luca M, Rega S, Ranieri B, Mauro C, Bossone E, Squire IB, Suzuki T, Marra AM. Biomarkers in Heart Failure: Clinical Insights. Heart Fail Clin 2021; 17:223-243. [PMID: 33673947 DOI: 10.1016/j.hfc.2021.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Andrea Salzano
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy.
| | - Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | | | - Mohamed Eltayeb
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Anna D'Agostino
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy
| | - Dennis Bernieh
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Mariarosaria De Luca
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Salvatore Rega
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Brigida Ranieri
- IRCCS SDN Nuclear and Diagnostic Research Institute, Naples, Italy
| | - Ciro Mauro
- AORN A Cardarelli, Cardiac Rehabilitation Unit, Naples, Italy
| | - Eduardo Bossone
- AORN A Cardarelli, Cardiac Rehabilitation Unit, Naples, Italy
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Alberto M Marra
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
106
|
Zhang Q, Hu M, Ma S. Association of Soluble Suppression of Tumorigenicity with No-Reflow Phenomenon and Long-Term Prognosis in Patients with Non-ST-Segment Elevation Acute Coronary Syndrome after Percutaneous Coronary Intervention. J Atheroscler Thromb 2021; 28:1289-1297. [PMID: 33551392 PMCID: PMC8629709 DOI: 10.5551/jat.59832] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aims:
Soluble suppression of tumorigenicity 2 (sST2) was validated to independently predict prognosis for heart failure (HF) and ST-segment elevation myocardial infarction (STEMI). In this study, we aimed to evaluate the relation between sST2 and coronary artery stenosis, and no-reflow phenomenon and one-year prognosis in patients with non-ST-segment elevation acute coronary syndrome (NSTE-ACS).
Methods:
This prospective study consecutively enrolled 205 patients who were diagnosed with NSTE-ACS and underwent percutaneous coronary intervention (PCI). sST2 was measured for all patients during admission. Patients were divided into two groups based on the optimal cutoff value: sST2 >34.2 ng/ml and sST2 ≤ 34.2 ng/ml groups.
Results:
Patients in the sST2 >34.2 ng/ml group was associated with higher Gensini scores and multivessel disease. sST2 had weak predictive value for no-reflow phenomenon (area under the curve [AUC], 0.662; 95% confidence interval [CI], 0.53–0.79;
P
=0.015) with 66.7% sensitivity and 65.2% specificity, and it also had independent predictive value of no-reflow phenomenon after adjusting for confounding factors (odds ratio [OR], 3.802; 95% CI, 1.03–14.11;
P
=0.046). sST2 >34.2 ng/ml had a commendable predictive value for the one-year prognosis (AUC, 0.84; 95% CI, 0.75–0.93;
P
<0.001) with 72% sensitivity and 84% specificity, and it independently predicted one-year major cardiovascular and cerebrovascular events (MACCE) (hazard ratio [HR], 10.22; 95% CI, 4.05–25.7;
P
<0.001).
Conclusion:
The sST2 concentration on admission is correlated with the degree of coronary artery stenosis. sST2 can predict both no-reflow and MACCE in patients with NSTE-ACS after PCI and was an independent predictor of MACCE and no-reflow phenomenon.
Collapse
Affiliation(s)
- Qinyao Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University
| | - Meirong Hu
- Department of Cardiology, Shengjing Hospital of China Medical University
| | - Shumei Ma
- Department of Cardiology, Shengjing Hospital of China Medical University
| |
Collapse
|
107
|
Sinha A, Gupta DK, Yancy CW, Shah SJ, Rasmussen-Torvik LJ, McNally EM, Greenland P, Lloyd-Jones DM, Khan SS. Risk-Based Approach for the Prediction and Prevention of Heart Failure. Circ Heart Fail 2021; 14:e007761. [PMID: 33535771 DOI: 10.1161/circheartfailure.120.007761] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Targeted prevention of heart failure (HF) remains a critical need given the high prevalence of HF morbidity and mortality. Similar to risk-based prevention of atherosclerotic cardiovascular disease, optimal HF prevention strategies should include quantification of risk in the individual patient. In this review, we discuss incorporation of a quantitative risk-based approach into the existing HF staging landscape and the clinical opportunity that exists to translate available data on risk estimation to help guide personalized decision making. We first summarize the recent development of key HF risk prediction tools that can be applied broadly at a population level to estimate risk of incident HF. Next, we provide an in-depth description of the clinical utility of biomarkers to personalize risk estimation in select patients at the highest risk of developing HF. We also discuss integration of genomics-enhanced approaches (eg, Titin [TTN]) and other risk-enhancing features to reclassify risk with a precision medicine approach to HF prevention. Although sequential testing is very likely to identify low and high-risk individuals with excellent accuracy, whether or not interventions based on these risk models prevent HF in clinical practice requires prompt attention including randomized placebo-controlled trials of candidate therapies in risk-enriched populations. We conclude with a summary of unanswered questions and gaps in evidence that must be addressed to move the field of HF risk assessment forward.
Collapse
Affiliation(s)
- Arjun Sinha
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL.,Department of Preventive Medicine, Feinberg School of Medicine (A.S., L.J.R.-T., P.G., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Deepak K Gupta
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN (D.K.G.)
| | - Clyde W Yancy
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Feinberg School of Medicine (A.S., L.J.R.-T., P.G., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Elizabeth M McNally
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Philip Greenland
- Department of Preventive Medicine, Feinberg School of Medicine (A.S., L.J.R.-T., P.G., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Donald M Lloyd-Jones
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL.,Department of Preventive Medicine, Feinberg School of Medicine (A.S., L.J.R.-T., P.G., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| | - Sadiya S Khan
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine (A.S., C.W.Y., S.J.S., E.M.N., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL.,Department of Preventive Medicine, Feinberg School of Medicine (A.S., L.J.R.-T., P.G., D.M.L.-J., S.S.K.), Northwestern University, Chicago, IL
| |
Collapse
|
108
|
Perrone MA, Storti S, Salvadori S, Pecori A, Bernardini S, Romeo F, Guccione P, Clerico A. Cardiac troponins: are there any differences between T and I? J Cardiovasc Med (Hagerstown) 2021; 22:797-805. [PMID: 33399346 DOI: 10.2459/jcm.0000000000001155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The most recent international guidelines recommend the measurement of cardiac troponin I (cTnI) and cardiac troponin T (cTnT) using high-sensitivity methods (hs-cTn) for the detection of myocardial injury and the differential diagnosis of acute coronary syndromes. Myocardial injury is a prerequisite for the diagnosis of acute myocardial infarction, but also a distinct entity. The 2018 Fourth Universal Definition of Myocardial Infarction states that myocardial injury is detected when at least one value above the 99th percentile upper reference limit is measured in a patient with high-sensitivity methods for cTnI or cTnT. Not infrequently, increased hs-cTnT levels are reported in patients with congenital or chronic neuromuscular diseases, while the hs-cTnI values are often in the normal range. Furthermore, some discrepancies between the results of laboratory tests for the two troponins are occasionally found in individuals apparently free of cardiac diseases, and also in patients with cardiac diseases. In this review article, authors discuss the biochemical, pathophysiological and analytical mechanisms which may cause discrepancies between hs-cTnI and hs-cTnT test results.
Collapse
Affiliation(s)
- Marco A Perrone
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital IRCCS Division of Cardiology, University of Rome Tor Vergata, Rome CNR-Regione Toscana G. Monasterio Foundation, Heart Hospital, Massa, and Scuola Superiore Sant'Anna CNR Institute of Clinical Physiology, Pisa Division of Clinical Biochemistry and Clinical Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Narayan V, Thompson EW, Demissei B, Ho JE, Januzzi JL, Ky B. Mechanistic Biomarkers Informative of Both Cancer and Cardiovascular Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 75:2726-2737. [PMID: 32466889 DOI: 10.1016/j.jacc.2020.03.067] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/03/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) and cancer are leading causes of morbidity and mortality worldwide. Although conventionally managed as separate disease processes, recent research has lent insight into compelling commonalities between CVD and cancer, including shared mechanisms for disease development and progression. In this review, the authors discuss several pathophysiological processes common to both CVD and cancer, such as inflammation, resistance to cell death, cellular proliferation, neurohormonal stress, angiogenesis, and genomic instability, in an effort to understand common mechanisms of both disease states. In particular, the authors highlight key circulating and genomic biomarkers associated with each of these processes, as well as their associations with risk and prognosis in both cancer and CVD. The purpose of this state-of-the-art review is to further our understanding of the potential mechanisms underlying cancer and CVD by contextualizing pathways and biomarkers common to both diseases.
Collapse
Affiliation(s)
- Vivek Narayan
- Division of Hematology/Medical Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth W Thompson
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Biniyam Demissei
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer E Ho
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Baim Institute for Clinical Research, Boston, Massachusetts
| | - Bonnie Ky
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
110
|
Gottdiener JS, Seliger S, deFilippi C, Christenson R, Baldridge AS, Kizer JR, Psaty BM, Shah SJ. Relation of Biomarkers of Cardiac Injury, Stress, and Fibrosis With Cardiac Mechanics in Patients ≥ 65 Years of Age. Am J Cardiol 2020; 136:156-163. [PMID: 32946864 DOI: 10.1016/j.amjcard.2020.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 01/16/2023]
Abstract
High sensitivity cardiac troponin T (hscTnT), soluble ST2 (sST2), N-terminal B-type natriuretic peptide (NT-proBNP), and galectin-3 are biomarkers of cardiac injury, stress, myocardial stretch, and fibrosis. Elevated levels are associated with poor outcomes. However, their association with cardiac mechanics in older persons is unknown. Associations between these biomarkers and cardiac mechanics derived from speckle tracking echocardiography, including left ventricular longitudinal strain (LVLS), early diastolic strain, and left atrial reservoir strain (LARS) were evaluated using standardized beta coefficients () in a cross sectional analysis with cardiac biomarkers in older patients without cardiovascular disease, low ejection fraction, or wall motion abnormalities. Biomarker associations with strain were attenuated by demographics and risk factors. In adjusted models, LVLS was associated with continuous measures of hscTnT (β∧-0.06, p = 0.020), sST2 (β∧ -0.05, p = 0.024) and NT-proBNP (β∧ -0.06, p = 0.007). "High" levels (i.e., greater than prognostic cutpoint) of hscTnT (>13 ng/ml), sST2 (>35 ng/ml), and NT-proBNP (>190 pg/ml) were also associated with worse LVLS. In risk factor adjusted models, LARS was associated with hscTnT (β∧ -0.08, p = 0.003) and NT-proBNP (β∧-0.18, p <0.0001). High hscTnT (>13 ng/ml) and high NT-proBNP (>190 pg/ml) were also both associated with worse LARS. Gal-3 was not associated with any strain measure. In conclusion, in persons ≥ 65 years of age, without cardiovascular disease, low ejection fraction, or wall motion abnormalities, hscTnT, sST2, and NT-proBNP are associated with worse LVLS. HscTnT and NT-proBNP are associated with worse LARS. In conclusion, these subclinical increases in blood biomarkers, and their associations with subtle diastolic and systolic dysfunction, may represent pre-clinical heart failure.
Collapse
Affiliation(s)
- John S Gottdiener
- Divisions of Cardiology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.
| | - Stephen Seliger
- Divisions of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Robert Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Abigail S Baldridge
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jorge R Kizer
- Division of Cardiology, Department of Medicine, UCSF School of Medicine, San Francisco, California; San Francisco VA Medical Center, San Francisco, California
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington and Kaiser Permanente Washington Health Research Institute, Seattle Washington
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
111
|
Application of Differential Network Enrichment Analysis for Deciphering Metabolic Alterations. Metabolites 2020; 10:metabo10120479. [PMID: 33255384 PMCID: PMC7761243 DOI: 10.3390/metabo10120479] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/11/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Modern analytical methods allow for the simultaneous detection of hundreds of metabolites, generating increasingly large and complex data sets. The analysis of metabolomics data is a multi-step process that involves data processing and normalization, followed by statistical analysis. One of the biggest challenges in metabolomics is linking alterations in metabolite levels to specific biological processes that are disrupted, contributing to the development of disease or reflecting the disease state. A common approach to accomplishing this goal involves pathway mapping and enrichment analysis, which assesses the relative importance of predefined metabolic pathways or other biological categories. However, traditional knowledge-based enrichment analysis has limitations when it comes to the analysis of metabolomics and lipidomics data. We present a Java-based, user-friendly bioinformatics tool named Filigree that provides a primarily data-driven alternative to the existing knowledge-based enrichment analysis methods. Filigree is based on our previously published differential network enrichment analysis (DNEA) methodology. To demonstrate the utility of the tool, we applied it to previously published studies analyzing the metabolome in the context of metabolic disorders (type 1 and 2 diabetes) and the maternal and infant lipidome during pregnancy.
Collapse
|
112
|
Tun B, Ehrbar R, Short M, Cheng S, Vasan RS, Xanthakis V. Association of Exhaled Carbon Monoxide With Ideal Cardiovascular Health, Circulating Biomarkers, and Incidence of Heart Failure in the Framingham Offspring Study. J Am Heart Assoc 2020; 9:e016762. [PMID: 33100134 PMCID: PMC7763395 DOI: 10.1161/jaha.120.016762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Exhaled carbon monoxide (eCO) is directly associated with traditional cardiovascular disease risk factors and incident cardiovascular disease. However, its relation with the cardiovascular health score and incidence of heart failure (HF) has not been investigated. Methods and Results We measured eCO in 3521 Framingham Heart Study Offspring participants attending examination cycle 6 (mean age 59 years, 53% women). We related the cardiovascular health score (composite of blood pressure, fasting plasma glucose, total cholesterol, body mass index, smoking, diet, and physical activity) to eCO adjusting for age, sex, and smoking. Higher cardiovascular health scores were associated with lower eCO (β=-0.02, P<0.0001), even among nonsmokers. Additionally, C-reactive protein, plasminogen activator inhibitor-1, fibrinogen, growth differentiation factor-15, homocysteine, and asymmetrical dimethylarginine were positively associated with eCO (P≤0.003 for all). The age- and sex-adjusted and multivariable-adjusted heritabilities of eCO were 49.5% and 31.4%, respectively. Over a median follow-up of 18 years, 309 participants (45% women) developed HF. After multivariable adjustment, higher eCO was associated with higher risk of HF (hazards ratio per SD increment: 1.39; 95% CI, 1.19-1.62 [P<0.001]) and with higher risk of HF with reduced ejection fraction (N=144 events; hazard ratio per SD increment in eCO: 1.43; 95% CI, 1.15-1.77 [P=0.001]). Conclusions In our community-based sample, higher levels of eCO were associated with lower cardiovascular health scores, an adverse cardiovascular biomarker profile, and a higher risk of HF, specifically HF with reduced ejection fraction. Our findings suggest that carbon monoxide may identify a novel pathway to HF development.
Collapse
Affiliation(s)
- Bradley Tun
- Department of Medicine Boston University School of Medicine and Boston Medical Center Boston MA
| | - Rachel Ehrbar
- Department of Biostatistics Boston University School of Public Health Boston MA
| | - Meghan Short
- Glenn Biggs Institute for Alzheimer's Disease & Neurodegenerative Diseases University of Texas Health Sciences Center San Antonio TX
| | - Susan Cheng
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Ramachandran S Vasan
- National Heart, Blood and Lung Institute Framingham Heart Study Framingham MA.,Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine Department of Medicine Boston University School of Medicine Boston MA.,Department of Epidemiology Boston University School of Public Health Boston MA
| | - Vanessa Xanthakis
- Department of Biostatistics Boston University School of Public Health Boston MA.,National Heart, Blood and Lung Institute Framingham Heart Study Framingham MA.,Sections of Preventive Medicine and Epidemiology and Cardiovascular Medicine Department of Medicine Boston University School of Medicine Boston MA
| |
Collapse
|
113
|
Wang Z, Yang F, Ma M, Bao Q, Shen J, Ye F, Xie X. The impact of growth differentiation factor 15 on the risk of cardiovascular diseases: two-sample Mendelian randomization study. BMC Cardiovasc Disord 2020; 20:462. [PMID: 33115406 PMCID: PMC7594331 DOI: 10.1186/s12872-020-01744-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Growth differentiation factor 15 (GDF-15), a stress responsive cytokine, belongs to transforming growth factor β cytokine superfamily. Some evidence support that it's involved in inflammation, coagulation, oxidative stress, endothelial dysfunction, and hemostasis. However, it's still controversial whether GDF-15 directly contributes to the morbidity and mortality of patients suffered with cardiovascular disease (CVD). Besides prospective cohort study and randomized controlled trial, Mendelian randomization (MR) is a genetic epidemiological method that exploits genetic variants as unbiased proxies for modifiable to determine the causal relationships between exposures and health outcomes. Herein, we introduced a two-sample MR approach to evaluate the causal relationships of circulating GDF-15 levels with major CVDs incidence. METHODS Genetic instruments and summary statistics for two-sample MR analysis were obtained from 5 independent large genome-wide association studies (GWAS) to investigate the causal correlation between circulating GDF-15 levels and 9 CVDs, respectively. Conventional inverse variance weighted method was adopted to evaluate the causality of GDF-15 with different outcomes; weighted median and MR egger were used for sensitivity analyses. RESULTS Among 9 SNPs identified from 5 GWASs in 2.6 million individuals, 5 SNPs (rs1227731, rs3195944, rs17725099, rs888663, rs749451) coming from chromosome 19 and containing the PGPEP1 and GDF-15 genes were employed. Based on the instruments, circulating GDF-15 levels significantly linked to the increased risk of cardioembolic stroke, atrial fibrillation, coronary artery disease and myocardial infarction. However, no significant causal association was observed for circulating GDF-15 levels with the incidence of any ischemic stroke, large-artery atherosclerotic stroke, small vessel stroke, heart failure and nonischemic cardiomyopathy. CONCLUSIONS The MR study provides with genetic evidence for the causal relationship of circulating GDF-15 levels with the increased risk of cardioembolic stroke, atrial fibrillation, coronary artery disease and myocardial infarction, but not any ischemic stroke, large-artery atherosclerotic stroke, small vessel stroke, heart failure and nonischemic cardiomyopathy. It indicates that GDF-15 might be a promising biomarker or potential therapeutic target for some CVDs.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Fangkun Yang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Menghuai Ma
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Qinyi Bao
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jinlian Shen
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Feiming Ye
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xiaojie Xie
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
114
|
Hinton J, Gabara L, Curzen N. Is the true clinical value of high-sensitivity troponins as a biomarker of risk? The concept that detection of high-sensitivity troponin 'never means nothing'. Expert Rev Cardiovasc Ther 2020; 18:843-857. [PMID: 32966128 DOI: 10.1080/14779072.2020.1828063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION High-sensitivity troponin (hs-cTn) assays are central to the diagnosis of myocardial infarction (MI). Their increased sensitivity has facilitated rapid pathways for the exclusion of MI. However, hs-cTn is now more readily detectable in patients without symptoms typical of MI, in whom a degree of myocardial injury is assumed. Recently, the practice of using the 99th centile of hs-cTn as a working 'upper reference limit' has been challenged. There is increasing evidence that hs-cTn may provide useful prognostic information, regardless of any suspicion of MI, and as such these assays may have potential as a general biomarker for mortality. This raises the concept that detection of hs-cTn 'never means nothing.' AREAS COVERED In this review, we will evaluate the evidence for the use of hs-cTn assays outside their common clinical indication to rule out or diagnose acute MI. EXPERT OPINION The data presented suggest that hs-cTn testing may in the future have a generalized role as a biomarker of mortality risk and may be used less as a test for ruling in acute MI, but will remain a frontline test to exclude that diagnosis in ED. Further, the data suggest that the detection of hs-cTn 'never means nothing.'
Collapse
Affiliation(s)
- Jonathan Hinton
- Coronary Research Group, University Hospital Southampton NHS Foundation Trust , Southampton, UK.,Faculty of Medicine, University of Southampton , Southampton, UK
| | - Lavinia Gabara
- Coronary Research Group, University Hospital Southampton NHS Foundation Trust , Southampton, UK.,Faculty of Medicine, University of Southampton , Southampton, UK
| | - Nick Curzen
- Coronary Research Group, University Hospital Southampton NHS Foundation Trust , Southampton, UK.,Faculty of Medicine, University of Southampton , Southampton, UK
| |
Collapse
|
115
|
Growth Differentiation Factor-15 as a Potent Predictor of Long-Term Mortality among Subjects with Osteoarthritis. J Clin Med 2020; 9:jcm9103107. [PMID: 32993054 PMCID: PMC7650798 DOI: 10.3390/jcm9103107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 11/20/2022] Open
Abstract
Background: Subjects with osteoarthritis (OA) are at increased risk for cardiovascular (CV) and all-cause mortality. Whether biomarkers improve outcome prediction in these patients remains to be elucidated. We investigated the association between growth differentiation factor 15 (GDF-15), a novel stress-responsive cytokine, and long-term all-cause mortality among OA patients. Methods: Within the Ulm Osteoarthritis Study, GDF-15 has been measured in the serum of 636 subjects, who underwent hip or knee arthroplasty between 1995 and 1996 (median age 65 years). Results: During a median follow-up of 19.7 years, a total of 402 deaths occurred. GDF-15 was inversely associated with walking distance. Compared to the bottom quartile (Q), subjects within the top quartile of GDF-15 demonstrated a 2.69-fold increased risk of dying (hazard ratio (HR) (95% confidence interval (CI)) 2.69 (1.82–3.96) adjusted for age, sex, BMI, smoking status, localization of OA, diabetes, maximum walking distance, total cholesterol, and cystatin C. Further adjustment for NT-proBNP, troponin I, and hs-C-reactive protein did not change the results appreciably (HR (95%CI) 1.56 (1.07–2.28); 1.75 (1.21–2.55); 2.32 (1.55–3.47) for Q2, Q3, and Q4 respectively, p for trend < 0.001). Conclusions: In subjects with OA, GDF-15 represents a potent predictor of decreased survival over >20 years, independently of conventional CV risk factors, renal, cardiac, and inflammatory biomarkers as well as walking disability, previously associated with increased mortality and lower extremity OA.
Collapse
|
116
|
Hara A, Niwa M, Kanayama T, Noguchi K, Niwa A, Matsuo M, Kuroda T, Hatano Y, Okada H, Tomita H. Galectin-3: A Potential Prognostic and Diagnostic Marker for Heart Disease and Detection of Early Stage Pathology. Biomolecules 2020; 10:biom10091277. [PMID: 32899694 PMCID: PMC7565392 DOI: 10.3390/biom10091277] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
The use of molecular biomarkers for the early detection of heart disease, before their onset of symptoms, is an attractive novel approach. Ideal molecular biomarkers, those that are both sensitive and specific to heart disease, are likely to provide a much earlier diagnosis, thereby providing better treatment outcomes. Galectin-3 is expressed by various immune cells, including mast cells, histiocytes and macrophages, and plays an important role in diverse physiological functions. Since galectin-3 is readily expressed on the cell surface, and is readily secreted by injured and inflammatory cells, it has been suggested that cardiac galectin-3 could be a marker for cardiac disorders such as cardiac inflammation and fibrosis, depending on the specific pathogenesis. Thus, galectin-3 may be a novel candidate biomarker for the diagnosis, analysis and prognosis of various cardiac diseases, including heart failure. The goals of heart disease treatment are to prevent acute onset and to predict their occurrence by using the ideal molecular biomarkers. In this review, we discuss and summarize recent developments of galectin-3 as a next-generation molecular biomarker of heart disease. Furthermore, we describe how galectin-3 may be useful as a diagnostic marker for detecting the early stages of various heart diseases, which may contribute to improved early therapeutic interventions.
Collapse
Affiliation(s)
- Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
- Correspondence: ; Tel.: +81-58-230-6225
| | - Masayuki Niwa
- Medical Education Development Center, Gifu University School of Medicine, Gifu 501-1194, Japan;
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Kei Noguchi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Mikiko Matsuo
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Takahiro Kuroda
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan;
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; (T.K.); (K.N.); (A.N.); (M.M.); (T.K.); (Y.H.); (H.T.)
| |
Collapse
|
117
|
Zhang X, Wang S, Liu J, Wang Y, Cai H, Wang D, Fang S, Yu B. D-dimer and the incidence of heart failure and mortality after acute myocardial infarction. Heart 2020; 107:237-244. [PMID: 32788198 DOI: 10.1136/heartjnl-2020-316880] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE D-dimer might serve as a marker of thrombogenesis and a hypercoagulable state following plaque rupture. Few studies explore the association between baseline D-dimer levels and the incidence of heart failure (HF), all-cause mortality in an acute myocardial infarction (AMI) population. We aimed to explore this association. METHODS We enrolled 4504 consecutive patients with AMI with complete data in a prospective cohort study and explored the association of plasma D-dimer levels on admission and the incidence of HF, all-cause mortality. RESULTS Over a median follow-up of 1 year, 1112 (24.7%) patients developed in-hospital HF, 542 (16.7%) patients developed HF after hospitalisation and 233 (7.1%) patients died. After full adjustments for other relevant clinical covariates, patients with D-dimer values in quartile 3 (Q3) had 1.51 times (95% CI 1.12 to 2.04) and in Q4 had 1.49 times (95% CI 1.09 to 2.04) as high as the risk of HF after hospitalisation compared with patients in Q1. Patients with D-dimer values in Q4 had more than a twofold (HR 2.34; 95% CI 1.33 to 4.13) increased risk of death compared with patients in Q1 (p<0.001). But there was no association between D-dimer levels and in-hospital HF in the adjusted models. CONCLUSIONS D-dimer was found to be associated with the incidence of HF after hospitalisation and all-cause mortality in patients with AMI.
Collapse
Affiliation(s)
- Xiaoyuan Zhang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Shanjie Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jinxin Liu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yini Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hengxuan Cai
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Duolao Wang
- Liverpool School of Tropical Medicine, Liverpool, Liverpool, UK
| | - Shaohong Fang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Bo Yu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China .,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| |
Collapse
|
118
|
Association between GDF15, poverty and mortality in urban middle-aged African American and white adults. PLoS One 2020; 15:e0237059. [PMID: 32764826 PMCID: PMC7413478 DOI: 10.1371/journal.pone.0237059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Mortality disparities are influenced by race and poverty. There is limited information about whether poverty influences biologic markers of mortality risk. Emerging data suggests that growth differentiation factor 15 (GDF15) is associated with mortality; however, the interplay between GDF15, sociodemographic factors and mortality is not known. We sought to evaluate the interactions between GDF15 and sex, race and poverty status on mortality. Serum GDF15 was measured in 1036 African American and white middle-aged men and women above and below 125% of the Federal poverty status from the Healthy Aging in Neighborhoods of Diversity across the Life Span (HANDLS) study. Multivariable adjusted Cox regression models were used to assess the association between log-transformed GDF15 (logGDF15) and 12-year mortality outcomes (all-cause, cardiovascular- and cancer-specific outcomes) and interactions with sex, race and poverty status. Likelihood ratio tests were used to assess significance of the interaction terms. Median GDF15 was 655.2 pg/mL (IQR = 575.1). During 12.2 years of follow-up, 331 died of which 94 cardiovascular- and 87 were cancer-specific deaths. One unit of increase in logGDF15 was associated with a hazard ratio for all-cause mortality, cardiovascular- and cancer-specific mortality of 2.26 (95% confidence interval [CI], 1.94–2.64), 2.74 (95%CI, 2.06–3.63) and 1.41 (95%CI, 1.00–2.00), respectively. There was an interaction between logGDF15 and poverty status on all-cause mortality (p<0.05). The GDF15×poverty status interaction term improved model calibration for all-cause mortality. Our study provides the first evidence that the effect of elevated GDF15 on all-cause mortality is modified by poverty status.
Collapse
|
119
|
Lockhart SM, Saudek V, O’Rahilly S. GDF15: A Hormone Conveying Somatic Distress to the Brain. Endocr Rev 2020; 41:bnaa007. [PMID: 32310257 PMCID: PMC7299427 DOI: 10.1210/endrev/bnaa007] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/02/2020] [Indexed: 12/27/2022]
Abstract
GDF15 has recently gained scientific and translational prominence with the discovery that its receptor is a GFRAL-RET heterodimer of which GFRAL is expressed solely in the hindbrain. Activation of this receptor results in reduced food intake and loss of body weight and is perceived and recalled by animals as aversive. This information encourages a revised interpretation of the large body of previous research on the protein. GDF15 can be secreted by a wide variety of cell types in response to a broad range of stressors. We propose that central sensing of GDF15 via GFRAL-RET activation results in behaviors that facilitate the reduction of exposure to a noxious stimulus. The human trophoblast appears to have hijacked this signal, producing large amounts of GDF15 from early pregnancy. We speculate that this encourages avoidance of potential teratogens in pregnancy. Circulating GDF15 levels are elevated in a range of human disease states, including various forms of cachexia, and GDF15-GFRAL antagonism is emerging as a therapeutic strategy for anorexia/cachexia syndromes. Metformin elevates circulating GDF15 chronically in humans and the weight loss caused by this drug appears to be dependent on the rise in GDF15. This supports the concept that chronic activation of the GDF15-GFRAL axis has efficacy as an antiobesity agent. In this review, we examine the science of GDF15 since its identification in 1997 with our interpretation of this body of work now being assisted by a clear understanding of its highly selective central site of action.
Collapse
Affiliation(s)
- Samuel M Lockhart
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Vladimir Saudek
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stephen O’Rahilly
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
120
|
Patient Management in Aortic Stenosis: Towards Precision Medicine Through Protein Analysis, Imaging and Diagnostic Tests. J Clin Med 2020; 9:jcm9082421. [PMID: 32731585 PMCID: PMC7463596 DOI: 10.3390/jcm9082421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 01/12/2023] Open
Abstract
Aortic stenosis is the most frequent valvular disease in developed countries. It progresses from mild fibrocalcific leaflet changes to a more severe leaflet calcification at the end stages of the disease. Unfortunately, symptoms of aortic stenosis are unspecific and only appear when it is too late, complicating patients' management. The global impact of aortic stenosis is increasing due to the growing elderly population. The disease supposes a great challenge because of the multiple comorbidities of these patients. Nowadays, the only effective treatment is valve replacement, which has a high cost in both social and economic terms. For that reason, it is crucial to find potential diagnostic, prognostic and therapeutic indicators that could help us to detect this disease in its earliest stages. In this article, we comprehensively review several key observations and translational studies related to protein markers that are promising for being implemented in the clinical field as well as a discussion about the role of precision medicine in aortic stenosis.
Collapse
|
121
|
Evidence on clinical relevance of cardiovascular risk evaluation in the general population using cardio-specific biomarkers. ACTA ACUST UNITED AC 2020; 59:79-90. [DOI: 10.1515/cclm-2020-0310] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/22/2020] [Indexed: 12/25/2022]
Abstract
Abstract
In recent years, the formulation of some immunoassays with high-sensitivity analytical performance allowed the accurate measurement of cardiac troponin I (cTnI) and T (cTnT) levels in reference subjects. Several studies have demonstrated the association between the risk of major cardiovascular events and cardiac troponin concentrations even for biomarker values within the reference intervals. High-sensitivity cTnI and cTnT methods (hs-cTn) enable to monitor myocardial renewal and remodelling, and to promptly identify patients at highest risk ofheart failure. An early and effective treatment of individuals at higher cardiovascular risk may revert the initial myocardial remodelling and slow down heart failure progression. Specific clinical trials should be carried out to demonstrate the efficacy and efficiency of the general population screening by means of cost-benefit analysis, in order to better identify individuals at higher risk for heart failure (HF) progression with hs-cTn methods.
Collapse
|
122
|
Kuster N, Huet F, Dupuy AM, Akodad M, Battistella P, Agullo A, Leclercq F, Kalmanovich E, Meilhac A, Aguilhon S, Cristol JP, Roubille F. Multimarker approach including CRP, sST2 and GDF-15 for prognostic stratification in stable heart failure. ESC Heart Fail 2020; 7:2230-2239. [PMID: 32649062 PMCID: PMC7524044 DOI: 10.1002/ehf2.12680] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Aims Inflammation and cardiac remodelling are common and synergistic pathways in heart failure (HF). Emerging biomarkers such as soluble suppression of tumorigenicity 2 (sST2) and growth differentiation factor‐15 (GDF‐15), which are linked to inflammation and fibrosis process, have been proposed as prognosis factors. However, their potential additive values remain poorly investigated. Methods and results Here, we aimed at evaluating inflammatory and remodelling biomarkers to predict both short‐term and long‐term mortality in a population with chronic HF in comparison with other classical clinical or biological markers (i.e. N terminal pro brain natriuretic peptide, hs‐cTnT, C‐reactive protein) alone or using meta‐analysis global group in chronic HF risk score in a cohort of 182 patients followed during 80 months (interquartile range: 12.3–90.0). Proportional hazard assumption does not hold for sST2 and C‐reactive protein, and follow‐up was split into short term (less than 1 year), midterm (between 1 and 5 years), and long term (after 5 years). In univariate analysis, C‐reactive protein and sST2 were predictive of short‐term mortality but not of middle term and long term whereas GDF‐15 was predictive of short and mid‐term but not of long‐term mortality. In a multivariate model after adjustment for meta‐analysis global group in chronic HF score including the three markers, only sST2 was predictive of short‐term mortality (P = 0.0225), and only GDF‐15 was predictive of middle term mortality (P = 0.0375). None of the markers was predictive of long‐term mortality. Conclusions Our results demonstrate that both sST2 and GDF‐15 significantly improve the prognosis evaluation of HF patients and suggest that the value of GDF‐15 is more sustained overtime and could predict middle term events.
Collapse
Affiliation(s)
- Nils Kuster
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Anne-Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France
| | - Mariama Akodad
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Pascal Battistella
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Audrey Agullo
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Florence Leclercq
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Eran Kalmanovich
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Alexandra Meilhac
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Francois Roubille
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| |
Collapse
|
123
|
Velagaleti RS, Larson MG, Enserro D, Song RJ, Vasan RS. Clinical course after a first episode of heart failure: insights from the Framingham Heart Study. Eur J Heart Fail 2020; 22:1768-1776. [DOI: 10.1002/ejhf.1918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/21/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Raghava S. Velagaleti
- Framingham Heart Study Framingham MA USA
- Cardiology Section, Department of Medicine Boston VA Healthcare System West Roxbury MA USA
| | - Martin G. Larson
- Framingham Heart Study Framingham MA USA
- Department of Mathematics and Statistics Boston University Boston MA USA
| | - Danielle Enserro
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics Roswell Park Comprehensive Cancer Center Buffalo NY USA
| | - Rebecca J. Song
- Department of Epidemiology Boston University School of Public Health Boston MA USA
| | - Ramachandran S. Vasan
- Framingham Heart Study Framingham MA USA
- Preventive Medicine and Cardiology Sections, Department of Medicine, School of Medicine, and Department of Epidemiology, School of Public Health Boston University Boston MA USA
| |
Collapse
|
124
|
Association of Lower Plasma Homoarginine Concentrations with Greater Risk of All-Cause Mortality in the Community: The Framingham Offspring Study. J Clin Med 2020; 9:jcm9062016. [PMID: 32604958 PMCID: PMC7356383 DOI: 10.3390/jcm9062016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/03/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
Lower circulating homoarginine concentrations have been associated with morbidity and mortality in patients with established cardiovascular disease (CVD). We assayed plasma homoarginine concentrations in 3331 Framingham Offspring Study participants attending examination cycle six (mean age 58.6 years, 53% women). We evaluated correlates of plasma homoarginine and related homoarginine to incident CVD and death. We also classified participants as having higher (upper quartile) versus lower (lower three quartiles) homoarginine and previously assayed asymmetric dimethylarginine (ADMA) concentrations, and created cross-classification groups. We observed 630 incident CVD events and 940 deaths during a median follow-up of 18 years. In multivariable regression analysis, homoarginine was associated positively with male sex, body mass index, anti-hypertensive medication use and systolic blood pressure, but inversely with age and smoking. Higher homoarginine levels were associated with a lower mortality risk (hazard ratio (HR) per SD increment, 0.83, 95% CI: 0.74–0.93) adjusting for standard CVD risk factors, and ADMA. Among the cross-classification groups, participants with higher homoarginine and lower ADMA had a lower mortality risk (HR, 0.81, 95% CI: 0.67–0.98) compared to those with low levels of both. Further studies are needed to dissect the mechanisms of the association of homoarginine and mortality over decades in the community.
Collapse
|
125
|
Aimo A, Januzzi JL, Bayes-Genis A, Vergaro G, Sciarrone P, Passino C, Emdin M. Clinical and Prognostic Significance of sST2 in Heart Failure: JACC Review Topic of the Week. J Am Coll Cardiol 2020; 74:2193-2203. [PMID: 31648713 DOI: 10.1016/j.jacc.2019.08.1039] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/07/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
Soluble suppression of tumorigenesis-2 (sST2) is released in response to vascular congestion and inflammatory and pro-fibrotic stimuli, and is a strong, independent predictor of mortality and heart failure (HF) hospitalization in patients with acute or chronic HF. sST2 meets 2 fundamental criteria for clinically useful biomarkers: accurate, repeated measurements are available at a reasonable cost, and the biomarker provides information not already available from a careful clinical assessment. In particular, the prognostic value of sST2 is additive to natriuretic peptides and (in the case of chronic HF) to high-sensitivity troponin T. Nevertheless, the need for a multibiomarker approach to risk stratification and the role of sST2 as a guide to therapy decision-making remain to be established. Four years after a consensus document on sST2, and following major advances in the comprehension of the clinical value of this biomarker, the authors felt it worthwhile to reappraise current knowledge on sST2 in HF.
Collapse
Affiliation(s)
- Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, Massachusetts
| | - Antoni Bayes-Genis
- Hospital Universitari Germans Trias i Pujol, Badalona (Barcelona), CIBERCV, Barcelona, Spain
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Paolo Sciarrone
- Cardiology Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Department, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| |
Collapse
|
126
|
Andersson J, Fall T, Delicano R, Wennberg P, Jansson JH. GDF-15 is associated with sudden cardiac death due to incident myocardial infarction. Resuscitation 2020; 152:165-169. [PMID: 32422242 DOI: 10.1016/j.resuscitation.2020.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 11/27/2022]
Abstract
AIMS Preventing sudden cardiac death (SCD) due to acute myocardial infarction (MI) in previously healthy patients is challenging. Proteomic analysis may lead to an understanding of biological mechanisms and provide predictive biomarkers. METHODS In this prospective, nested case-control study from northern Sweden, 87 candidate cardiovascular protein biomarkers were studied in 244 individuals who later died within 24 h from an incident MI and 244 referents without MI and individually matched for age, sex and date of health examination and alive at the date of event in the index person. Association analysis was conducted using conditional logistic regression. Bonferroni correction was applied to avoid false positive findings. RESULTS Ten proteins were associated with future SCD due to acute MI in the non-adjusted analysis. The strongest association were found for growth differentiation factor 15 (GDF-15) with an odds ratio (OR) of 1.79 (95% confidence interval [CI] 1.41, 2.25) per standard deviation increase in protein, and urokinase-type plasminogen activator receptor with an OR of 1.66 (95% CI 1.34, 2.06). In models adjusted for lipid levels, body mass index, education, smoking, hypertension and C-reactive protein, only association with GDF-15 remained (OR 1.47 (95% 1.11, 1.95)). CONCLUSION Elevated levels of GDF-15 are associated with increased risk of SCD within 24 h of incident MI. Further research may enable the use of GDF-15 together with other clinical and biological markers to guide primary preventive interventions for individuals at high risk for SCD.
Collapse
Affiliation(s)
- Jonas Andersson
- Department of Public Health and Clinical Medicine, Skellefteå Research Unit, Umeå University, Sweden.
| | - Tove Fall
- Uppsala University and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Rachel Delicano
- Uppsala University and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Sweden
| | - Jan-Håkan Jansson
- Department of Public Health and Clinical Medicine, Skellefteå Research Unit, Umeå University, Sweden
| |
Collapse
|
127
|
|
128
|
Sarhene M, Wang Y, Wei J, Huang Y, Li M, Li L, Acheampong E, Zhengcan Z, Xiaoyan Q, Yunsheng X, Jingyuan M, Xiumei G, Guanwei F. Biomarkers in heart failure: the past, current and future. Heart Fail Rev 2020; 24:867-903. [PMID: 31183637 DOI: 10.1007/s10741-019-09807-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the enhanced knowledge of the pathophysiology of heart failure (HF), it still remains a serious syndrome with substantial morbidity, mortality, and frequent hospitalizations. These are due to the current improvements in other cardiovascular diseases (like myocardial infarction), the aging population, and growing prevalence of comorbidities. Biomarker-guided management has brought a new dimension in prognostication, diagnosis, and therapy options. Following the recommendation of natriuretic peptides (B-type natriuretic peptide and N-terminal-proBNP), many other biomarkers have been thoroughly studied to reflect different pathophysiological processes (such as fibrosis, inflammation, myocardial injury, and remodeling) in HF and some of them (like cardiac troponins, soluble suppression of tumorigenesis-2, and galectin 3) have subsequently been recommended to aid in the diagnosis and prognostication in HF. Consequently, multi-marker approach has also been approved owing to the varied nature of HF syndrome. In this review, we discussed the guidelines available for HF biomarkers, procedures for evaluating novel markers, and the utilities of both emerging and established biomarkers for risk stratification, diagnosis, and management of HF in the clinics. We later looked at how the rapidly emerging field-OMICs, can help transform HF biomarkers discoveries and establishment.
Collapse
Affiliation(s)
- Michael Sarhene
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Yili Wang
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Jing Wei
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Yuting Huang
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Min Li
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Lan Li
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China
| | - Enoch Acheampong
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhou Zhengcan
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qin Xiaoyan
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xu Yunsheng
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mao Jingyuan
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China
| | - Gao Xiumei
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fan Guanwei
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Number 314 Anshanxi Road, Nankai District, Tianjin, China. .,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
129
|
Fernandez C, Rysä J, Ström K, Nilsson J, Engström G, Orho-Melander M, Ruskoaho H, Melander O. Circulating protein biomarkers predict incident hypertensive heart failure independently of N-terminal pro-B-type natriuretic peptide levels. ESC Heart Fail 2020; 7:1891-1899. [PMID: 32410391 PMCID: PMC7373917 DOI: 10.1002/ehf2.12757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/28/2022] Open
Abstract
Aims Hypertension is the leading cause for the development of heart failure (HF). Here, we aimed to identify cardiomyocyte stretch‐induced circulating biomarkers for predicting hypertension‐associated HF. Methods and results Circulating levels of 149 proteins were measured by proximity extension assay at baseline examination in 4742 individuals from the Malmö Diet and Cancer study. Protein levels were compared with stretch‐activated gene expression changes in cultured neonatal rat ventricular myocytes (NRVMs) in response to 1–48 h of mechanical stretch. We also studied the association between protein levels and hypertension and HF incidence using respectively binary logistic and Cox regressions. Levels of 35 proteins were differentially expressed after Bonferroni correction in incident HF vs. control (P < 3.4E−4). Growth differentiation factor‐15 (GDF‐15), interleukin‐6 (IL‐6), IL‐1 receptor type 1, and urokinase plasminogen activator surface receptor had corresponding mRNA levels up‐regulated by stretch in NRVMs at all time points (P < 0.05). These four proteins were individually associated with increased risk of HF after age and sex adjustment [hazard ratio (HR) per standard deviation: 1.19 ≤ HR ≤ 1.49, P ≤ 4.90E−3]. GDF‐15 and IL‐6 were associated with HF independently of each other (1.22 ≤ HR ≤ 1.33, P ≤ 0.001). In subjects with hypertension, these associations remained significant after further adjustment for N‐terminal pro‐B‐type natriuretic peptide (NT‐proBNP) levels (1.23 ≤ HR ≤ 1.45, P ≤ 0.001). A higher fasting value of a GDF‐15, IL‐6 score aggregate was associated with increased risk of hypertensive HF after adjustment for all traditional risk factors for HF and NT‐proBNP (HR = 1.31, P = 2.19E−4). Conclusions Cardiomyocyte mRNA levels of GDF‐15 and IL‐6 are consistently up‐regulated by stretch, and their circulating protein levels predict HF in hypertensive subjects independently of NT‐proBNP during long‐term follow‐up. Our results encourage further studies on lower blood pressure goals in hypertensive subjects with high GDF‐15 and IL‐6, and interventions targeted at stretch‐induced cardiomyocyte expressed biomarkers.
Collapse
Affiliation(s)
- Celine Fernandez
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kristoffer Ström
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | | | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Olle Melander
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| |
Collapse
|
130
|
Stein NR, Zelnick LR, Anderson AH, Christenson RH, deFilippi CR, Deo R, Go AS, He J, Ky B, Lash JP, Seliger SL, Soliman EZ, Shlipak MG, Bansal N. Associations Between Cardiac Biomarkers and Cardiac Structure and Function in CKD. Kidney Int Rep 2020; 5:1052-1060. [PMID: 32647762 PMCID: PMC7335964 DOI: 10.1016/j.ekir.2020.04.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Subclinical changes to cardiac structure and function detected with echocardiography precede the development of clinical heart failure (HF) in persons with chronic kidney disease (CKD). Circulating cardiac biomarkers may reflect these pathophysiological changes. This study investigated associations between established biomarkers (N-terminal pro-B-type natriuretic peptide [NT-proBNP] and high-sensitivity troponin T [hsTnT]) and novel biomarkers (growth differentiation factor 15 [GDF-15], galectin-3 [Gal-3], and soluble ST-2 [sST-2]), using echocardiographic measurements in persons with CKD. Methods In cross-sectional analyses among 2101 participants with mild to moderate CKD in the Chronic Renal Insufficiency Cohort (CRIC), biomarker levels measured at baseline were evaluated with echocardiographic measurements 1 year later. These included left ventricular mass index (LVMI), left ventricular end-systolic volume (LVESV), left ventricular end-diastolic volume (LVEDV), left ventricular ejection fraction (LVEF), and left atrial diameter (LAD). Multivariable linear regression analyses tested associations of each biomarker with echocardiographic measurements, adjusting for covariates. Results GDF-15 was significantly associated with higher LVMI (1.0 g/m2.7; 95% CI, 0.4–1.7), LVESV (0.4 ml/m2.7; 95% CI, 0.0–0.7), and LVEDV (0.6 ml/m2.7; 95% CI, 0.1–1.1), but not with LVEF or LAD. These findings were not significant when adjusting for NT-proBNP and hsTnT. Gal-3 and sST-2 had no significant associations. Higher levels of NT-proBNP and hsTnT were associated with all echocardiographic measurements. Conclusion In patients with CKD, the novel biomarker GDF-15, a marker of inflammation and tissue injury, and clinical biomarkers NT-proBNP and hsTnT, were associated with echocardiographic measurements of subclinical cardiovascular disease. Collectively, these biomarkers may highlight biological pathways that contribute to the development of clinical HF.
Collapse
Affiliation(s)
- Nathan R Stein
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Leila R Zelnick
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Amanda H Anderson
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Robert H Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Rajat Deo
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan S Go
- Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Jiang He
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Bonnie Ky
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James P Lash
- Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Stephen L Seliger
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Elsayed Z Soliman
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael G Shlipak
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Nisha Bansal
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | | |
Collapse
|
131
|
Watson CJ, Tea I, O'Connell E, Glezeva N, Zhou S, James S, Gallagher J, Snider J, Januzzi JL, Ledwidge MT, McDonald KM. Comparison of longitudinal change in sST2 vs BNP to predict major adverse cardiovascular events in asymptomatic patients in the community. J Cell Mol Med 2020; 24:6495-6499. [PMID: 32347644 PMCID: PMC7294165 DOI: 10.1111/jcmm.15004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 11/27/2022] Open
Abstract
Biomarker‐based preventative and monitoring strategies are increasingly used for risk stratification in cardiovascular (CV) disease. The aim of this study was to investigate the utility of longitudinal change in B‐type natriuretic peptide (BNP) and sST2 concentrations for predicting incident major adverse CV events (MACE) (heart failure, myocardial infarction, arrhythmia, stroke/transient ischaemic attack and CV death) in asymptomatic community‐based patients with risk factors but without prevalent MACE at enrolment. The study population consisted of 282 patients selected from the longitudinal STOP‐HF study of asymptomatic patients with risk factors for development of MACE. Fifty‐two of these patients developed a MACE. The study was run in two phases comprising of an initial investigative cohort (n = 195), and a subsequent 2:1 (No MACE: MACE) propensity matched verification cohort (n = 87). BNP and sST2 were quantified in all patients at two time points a median of 2.5 years apart. Results highlighted that longitudinal change in sST2 was a statistically significant predictor of incident MACE, (AUC 0.60). A one‐unit increment in sST2 change from baseline to follow up corresponded to approximately 7.99% increase in the rate of one or more incident MACE, independent of the baseline or follow‐up concentration. In contrast, longitudinal change value of BNP was not associated with MACE. In conclusion, longitudinal change in sST2 but not BNP was associated with incident MACE in asymptomatic, initially event‐free patients in the community. Further work is required to evaluate the clinical utility of change in sST2 in risk prediction and event monitoring in this setting.
Collapse
Affiliation(s)
- Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland.,STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland.,UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Isaac Tea
- Internal Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| | - Eoin O'Connell
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland
| | - Nadezhda Glezeva
- UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Shuaiwei Zhou
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland
| | - Stephanie James
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland
| | - Joe Gallagher
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland.,UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | | | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Mark T Ledwidge
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland.,UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Ken M McDonald
- STOP-HF Group, St Vincent's University Hospital, Dublin, Ireland.,UCD Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
132
|
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in developed countries, and it is the primary cause of mortality in the elderly worldwide. The processes of inflammatory response activation, production and release of pro-inflammatory cytokines, activation of the complement system, synthesis of autoantibodies, and overexpression of Class II major histocompatibility complex molecules contribute to the HF development and progression. High levels of circulating cytokines correlate with the severity of HF, measured with the use of New York Heart Association's classification, and prognosis of the disease. In HF, there is an imbalance between pro-inflammatory and anti-inflammatory cytokines. Concentrations of several interleukins are increased in HF, including IL-1β, IL-6, IL-8, IL-9, IL-10, IL-13, IL-17, and IL-18, whereas the levels of IL-5, IL-7, or IL-33 are down-regulated. Concentrations of inflammatory mediators are associated with cardiac function and can be HF markers and predictors of adverse outcomes or mortality. This review presents the role of interleukins, which contribute to the HF initiation and progression, the importance of their pathways in transition from myocardial injury to HF, and the role of interleukins as markers of disease severity and outcome predictors.
Collapse
|
133
|
Zhang H, Dong P, Yang X, Du L, Wang K, Yan P, Zhang H, Wang T, Zhao X, Guo T. Prognostic indicators of new onset atrial fibrillation in patients with acute coronary syndrome. Clin Cardiol 2020; 43:647-651. [PMID: 32285941 PMCID: PMC7298978 DOI: 10.1002/clc.23363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 11/23/2022] Open
Abstract
Background This study aims to estimate prognostic indicators of new onset atrial fibrillation (AF) in patients with acute coronary syndrome (ACS) through 3 to 5 years of follow‐up. Hypothesis For patients with ACS, some prognostic indicators can be used to predict new onset AF. Methods The Improving Care for Cardiovascular Disease in China‐ACS (CCC‐ACS) program was launched in 2014 by a collaborative initiative of the American Heart Association and Chinese Society of Cardiology. We enrolled 866 patients with ACS in a telephone follow‐up program. We inquired about each patient's general health and invited each patient to our hospital for further consultation. We also performed ambulatory electrocardiography and other relevant examinations. Results A total of 743 ACS patients were included in the study. After 3 to 5 years, 50 (0.67%) patients developed AF. In multivariable Cox models adjusting for AF risk factors in ACS patients, we found that NT‐proBNP [hazard ratio (HR) 2.625, 1.654‐4.166, P < .05], creatine kinase‐MB (CK‐MB) (HR 4.279, 1.887‐9.703, P < .05), and left ventricular ejection fraction (LVEF) (HR 0.01, 0.001‐0.352, P < .05) were significantly associated with AF receiver operating characteristic (ROC) curves were used to determine a cutoff level for AF screening. NT‐proBNP using a cutoff of 1705 ng/L resulted in a sensitivity of 58% and a specificity of 89.8%. CK‐MB using a cutoff of 142.5 ng/L resulted in a sensitivity of 73.3% and a specificity of 58.3%. Conclusion For patients with ACS, NT‐proBNP, CK‐MB, and LVEF have a considerable prognostic value for predicting whether AF would be detected during follow‐up.
Collapse
Affiliation(s)
- Hengliang Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Pingshuan Dong
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xvming Yang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Laijing Du
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ke Wang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Peng Yan
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Huifeng Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Tengfei Wang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xikun Zhao
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Tengfei Guo
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
134
|
Velagaleti RS, Short MI, Larson MG, Vasan RS. Prognosis of "pre-heart failure" clinical phenotypes. PLoS One 2020; 15:e0231254. [PMID: 32275698 PMCID: PMC7147998 DOI: 10.1371/journal.pone.0231254] [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: 08/07/2019] [Accepted: 03/19/2020] [Indexed: 11/21/2022] Open
Abstract
Background Heart failure (HF) is a clinical syndrome where diagnostic certainty varies. The prognosis of individuals with some clinical features of HF, but without the fully overt syndrome, is unclear. Therefore, we sought to evaluate their natural history. Methods and results Between 1990 and 2009, all suspected HF cases in the Framingham Heart Study were adjudicated into 3 groups reflecting varying diagnostic certainty: definite (meeting HF diagnostic criteria; n = 479), possible (meeting HF criteria but with an alternative explanation for findings; n = 135), and probable (insufficient criteria for definite HF; n = 121) HF. Age-and-sex-matched individuals (n = 1112) without HF or cardiovascular disease (CVD) were controls. Using multivariable-adjusted Cox regression, we compared the possible/probable HF groups with controls regarding risk of incident definite HF, coronary heart disease (CHD), other CVD or death; and with definite HF regarding risk of latter three outcomes. During follow-up (mean 8.6 years), ~90% of individuals with possible, probable and definite HF experienced CVD events or died. Compared with controls, those with possible or probable HF experienced higher hazards for definite HF, CHD, other CVD and death (hazards ratios [HR] 1.35–9.31; p<0.05). The possible/probable groups did not differ from the definite HF group for risk of any outcome. Compared with the possible HF group, the probable HF group had a higher propensity for definite HF (HR 1.64, with a higher proportion of ischemic HF) but lower risk of death (HR 0.69). Conclusions Individuals meeting partial criteria for HF are at a substantial risk for progression to HF, CVD, and mortality.
Collapse
Affiliation(s)
- Raghava S. Velagaleti
- Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medicine, Cardiology Section, Boston VA Healthcare System, West Roxbury, Massachusetts, United States of America
- * E-mail:
| | - Meghan I. Short
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Martin G. Larson
- Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
| | - Ramachandran S. Vasan
- Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medicine, Preventive Medicine and Cardiology Sections, School of Medicine, and Department of Epidemiology, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
135
|
Krintus M, Braga F, Kozinski M, Borille S, Kubica J, Sypniewska G, Panteghini M. A study of biological and lifestyle factors, including within-subject variation, affecting concentrations of growth differentiation factor 15 in serum. Clin Chem Lab Med 2020; 57:1035-1043. [PMID: 30471215 DOI: 10.1515/cclm-2018-0908] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
Abstract
Background Growth differentiation factor 15 (GDF-15) is an emerging cardiovascular biomarker, and a fully automated immunoassay has recently become available. The objectives of the study were to identify biological and lifestyle factors affecting serum GDF-15 concentrations and derive robust reference intervals, and to estimate GDF-15 within-subject biological variation and derived indices. Methods A presumably healthy population of 533 questionnaire-screened adults was used to identify the biological and lifestyle determinants of serum GDF-15. Following stringent exclusion criteria, a final group of 173 individuals was selected to establish GDF-15 reference interval. Twenty-six healthy volunteers were enrolled in the biological variation substudy. Results Using a multiple regression model, age, B-type natriuretic peptide and C-reactive protein as well as smoking status were significantly related to serum GDF-15 concentrations. The upper reference limit (URL) for serum GDF-15 concentrations (90% confidence interval [CI]) was 866 ng/L (733-999 ng/L), with no sex-related difference. Although GDF-15 tended to increase with age, the weak dependence of marker from age does not justify age-related URL. The within-subject CV was 6.3% (95% CI, 4.5%-8.5%), with no sex difference in intraindividual variances. The reference change value (RCV) for GDF-15 was 23%, and two are the specimens required to ensure that the mean GDF-15 result is within ±10% of the individual's homeostatic set point. Conclusions By identifying the main factors influencing serum GDF-15 concentrations, we robustly established the URL to be applied in adult population. As intraindividual variation of GDF-15 is relatively low, monitoring longitudinal changes in its concentrations over time using RCV can be a good alternative for interpreting GDF-15 in clinical setting.
Collapse
Affiliation(s)
- Magdalena Krintus
- Department of Laboratory Medicine, Nicolaus Copernicus University, Collegium Medicum, 9 Sklodowskiej-Curie Street, 85-094 Bydgoszcz, Poland, Phone: +48 52 585 40 23, Fax: +48 52 585 40 24
| | - Federica Braga
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy.,Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Marek Kozinski
- Department of Principles of Clinical Medicine, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Simona Borille
- Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Grazyna Sypniewska
- Department of Laboratory Medicine, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Mauro Panteghini
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy.,Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
136
|
Soumagne T, Roche N, Guillien A, Bouhaddi M, Rocchi S, Hue S, Claudé F, Bizard L, Andujar P, Dalphin JC, Degano B. Cardiovascular Risk in COPD. Chest 2020; 157:834-845. [DOI: 10.1016/j.chest.2019.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 10/23/2019] [Accepted: 11/04/2019] [Indexed: 01/21/2023] Open
|
137
|
Growth differentiation factor-15 is a prognostic marker in patients with intermediate coronary artery disease. J Geriatr Cardiol 2020; 17:210-216. [PMID: 32362919 PMCID: PMC7189259 DOI: 10.11909/j.issn.1671-5411.2020.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Growth differentiation factor-15 (GDF-15) is involved in multiple processes that are associated with coronary artery disease (CAD). However, little is known about the association between GDF-15 and the future ischemic events in patients with intermediate CAD. This study was conducted to investigate whether plasma GDF-15 constituted risk biomarkers for future cardiovascular events in patients with intermediate CAD. METHODS A prospective study was performed based on 541 patients with intermediate CAD (20%-70%). GDF-15 of each patient was determined in a blinded manner. The primary endpoint was major adverse cardiac event (MACE), which was defined as a composite of all-cause death, nonfatal myocardial infarction, revascularization and readmission due to angina pectoris. RESULTS After a median follow-up of 64 months, 504 patients (93.2%) completed the follow-up. Overall, the combined endpoint of MACE appeared in 134 patients (26.6%) in the overall population: 26 patients died, 11 patients suffered a nonfatal myocardial infarction, 51 patients underwent revascularization, and 46 patients were readmitted for angina pectoris. The plasma levels of GDF-15 (median: 1172.02 vs. 965.25 pg/mL, P = 0.014) were higher in patients with ischemic events than those without events. After adjusting for traditional risk factors, higher GDF-15 levels were significantly associated with higher incidence of the composite endpoint of MACE (HR = 1.244, 95% CI: 1.048-1.478, Quartile 4 vs. Quartile 1, P = 0.013). CONCLUSIONS The higher level of GDF-15 was an independent predictor of long-term adverse cardiovascular events in patients with intermediate CAD.
Collapse
|
138
|
Suthahar N, Meems LMG, Ho JE, de Boer RA. Sex-related differences in contemporary biomarkers for heart failure: a review. Eur J Heart Fail 2020; 22:775-788. [PMID: 32220046 PMCID: PMC7319414 DOI: 10.1002/ejhf.1771] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/28/2022] Open
Abstract
The use of circulating biomarkers for heart failure (HF) is engrained in contemporary cardiovascular practice and provides objective information about various pathophysiological pathways associated with HF syndrome. However, biomarker profiles differ considerably among women and men. For instance, in the general population, markers of cardiac stretch (natriuretic peptides) and fibrosis (galectin‐3) are higher in women, whereas markers of cardiac injury (cardiac troponins) and inflammation (sST2) are higher in men. Such differences may reflect sex‐specific pathogenic processes associated with HF risk, but may also arise as a result of differences in sex hormone profiles and fat distribution. From a clinical perspective, sex‐related differences in biomarker levels may affect the objectivity of biomarkers in HF management because what is considered to be ‘normal’ in one sex may not be so in the other. The objectives of this review are, therefore: (i) to examine the sex‐specific dynamics of clinically relevant HF biomarkers in the general population, as well as in HF patients; (ii) to discuss the overlap between sex‐related and obesity‐related effects, and (iii) to identify knowledge gaps to stimulate research on sex‐related differences in
HF.
Collapse
Affiliation(s)
- Navin Suthahar
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Jennifer E Ho
- Division of Cardiology, Department of Medicine, and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rudolf A de Boer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| |
Collapse
|
139
|
Yan I, Börschel CS, Neumann JT, Sprünker NA, Makarova N, Kontto J, Kuulasmaa K, Salomaa V, Magnussen C, Iacoviello L, Di Castelnuovo A, Costanzo S, Linneberg A, Söderberg S, Zeller T, Ojeda-Echevarria FM, Blankenberg S, Westermann D. High-Sensitivity Cardiac Troponin I Levels and Prediction of Heart Failure: Results From the BiomarCaRE Consortium. JACC-HEART FAILURE 2020; 8:401-411. [PMID: 32171759 DOI: 10.1016/j.jchf.2019.12.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aims of this study were to characterize the association of high-sensitivity cardiac troponin I (hs-cTnI) with heart failure (HF), to determine its predictive value beyond classical cardiovascular risk factors (CVRFs) and N-terminal pro-B-type natriuretic peptide, and to derive a relevant cutoff for potential clinical application. BACKGROUND HF is an important contributor to the overall burden of cardiovascular disease. Early identification of individuals at risk could be beneficial for preventive therapies. METHODS Based on the Biomarker for Cardiovascular Risk Assessment in Europe consortium, we analyzed individual-level data from 4 prospective population-based cohort studies including 48,455 individuals. Participants with myocardial infarction, HF, and stroke at baseline were excluded. We investigated the value of adding hs-cTnI to CVRFs and N-terminal pro-B-type natriuretic peptide using Cox proportional hazards survival models and for prediction by calculating C-statistics and Brier score. RESULTS The median age of the study population was 51 years, and the median follow-up time for occurrence of HF was 6.61 years. Cox regression models adjusted for age, sex, and CVRFs revealed a significant association of hs-cTnI with incident HF (hazard ratio: 1.42 per log [ng/l] unit change [95% confidence interval: 1.31 to 1.53]). The best predictive value was achieved in the model with CVRFs (base model) and both biomarkers (C-index = 0.862; 95% confidence interval: 0.841 to 0.882). Optimal hs-cTnI cutoff values of 2.6 ng/l for women and 4.2 ng/l for men were derived for selecting individuals at risk. CONCLUSIONS In this large dataset from the general population, hs-cTnI could show its independence for the prognosis of HF.
Collapse
Affiliation(s)
- Isabell Yan
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany
| | - Christin S Börschel
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Johannes T Neumann
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Ngoc A Sprünker
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany
| | - Nataliya Makarova
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Jukka Kontto
- National Institute for Health and Welfare, Helsinki, Finland
| | - Kari Kuulasmaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Christina Magnussen
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy; Research Center in Epidemiology and Preventive Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Simona Costanzo
- Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Tanja Zeller
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | | | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany.
| |
Collapse
|
140
|
Abstract
PURPOSE OF REVIEW Heart failure represents a major growing health problem in developed world. This article aims to review recent heart failure trials that have significantly impacted the management of heart failure. RECENT FINDINGS Despite advances in heart failure, mortality and morbidity remains elevated amongst patients. Recent clinical trials demonstrate promising treatment strategies that likely impact clinical practice; including heart failure prevention with the use of SGLT2-inhibitors in patients with diabetes and cardiovascular risk, new treatments that may abrogate disease progression in cardiac amyloidosis, intravenous iron therapy in iron deficiency anemia in chronic systolic heart failure, predischarge treatment with angiotensin receptor blocker with neprilysin inhibition (ARNi) in patients hospitalized for acute decompensated heart failure, and newer continuous flow left ventricular assist device with increased durability and efficacy in patients with Stage D heart failure. SUMMARY Recent clinical trials with SGLT2 inhibitors, therapies targeting transthyretin cardiac amyloidosis, iron, angiotensin receptor blocker with neprilysin inhibition and newer mechanical circulatory support devices are very promising as practice changing new treatment strategies in prevention and treatment of heart failure. This article presents a summary of important trials and should be of practical value to both clinicians and researchers.
Collapse
|
141
|
Le Goff C, Farré Segura J, Dufour P, Kaux JF, Cavalier E. Intense sport practices and cardiac biomarkers. Clin Biochem 2020; 79:1-8. [PMID: 32097617 DOI: 10.1016/j.clinbiochem.2020.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022]
Abstract
Biomarkers are well established for the diagnosis of myocardial infarction, heart failure and cardiac fibrosis. Different papers on cardiac biomarker evolution during exercise have been published in the literature and generally show mild to moderate elevations. However, the mechanism responsible for these elevations, reflecting physiological or even pathophysiological changes, still has to be clearly elucidated. There are also indications of higher cardiac risk in poorly trained athletes than in well-trained athletes. Whether regular repetition of intensive exercise might lead, in the longer term, to fibrosis and heart failure remains to be determined. In this review, we summarized the main research about the effects of intense exercise (in particular, running) on cardiac biomarkers (including troponins, natriuretic peptides, etc.). We found that cardiac fibrosis biomarkers seemed to be the most informative regarding the biological impact of intense physical activity.
Collapse
Affiliation(s)
- C Le Goff
- Department of Clinical Chemistry, University Hospital of Liege, Belgium.
| | - J Farré Segura
- Department of Clinical Chemistry, University Hospital of Liege, Belgium
| | - P Dufour
- Department of Clinical Chemistry, University Hospital of Liege, Belgium
| | - J F Kaux
- Department of Clinical Chemistry, University Hospital of Liege, Belgium
| | - E Cavalier
- Department of Clinical Chemistry, University Hospital of Liege, Belgium
| |
Collapse
|
142
|
Short-Term Prognosis Value of sST2 for an Unfavorable Outcome in Hypertensive Patients. DISEASE MARKERS 2020; 2020:8143737. [PMID: 32089758 PMCID: PMC7026724 DOI: 10.1155/2020/8143737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022]
Abstract
Background sST2 represents a useful biomarker for the diagnosis and prognosis of patients with heart failure, but limited data is available on its role in patients with hypertension. The aim of this study is to evaluate the short-term prognosis value of sST2 for an unfavorable outcome in hypertensive patients. Methods This was a prospective observational study which enrolled 80 patients with hypertension, who were followed for one year. All patients underwent clinical, laboratory (including sST2), and echocardiographic assessment at baseline. The patients were grouped according to the cardiovascular (CV) events reported during the follow-up: group A (with CV events) and group B (without CV events). Results Overall, 59 CV events were reported during the follow-up period. Compared to group B, the patients in group A had significantly higher sST2 levels, a higher number of CV risk factors, and a higher left ventricle mass. Except for the diastolic dysfunction parameters, the echocardiographic findings were similar in the two groups. Patients in group A had a lower E/A ratio, larger deceleration time, and increased telediastolic pressure as quantified by the E/E/p = 0.006, Kaplan-Meier analysis). Conclusions sST2 levels were correlated with the risk of adverse CV outcomes in hypertensive patients and may represent a useful prognostic marker in these patients.
Collapse
|
143
|
Thorsteinsdottir H, Salvador CL, Mjøen G, Lie A, Sugulle M, Tøndel C, Brun A, Almaas R, Bjerre A. Growth Differentiation Factor 15 in Children with Chronic Kidney Disease and after Renal Transplantation. DISEASE MARKERS 2020; 2020:6162892. [PMID: 32089755 PMCID: PMC7026715 DOI: 10.1155/2020/6162892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 10/29/2019] [Accepted: 01/04/2020] [Indexed: 12/23/2022]
Abstract
Growth differentiation factor 15 (GDF-15) is strongly associated with cardiovascular disease (CVD). The aim of our study was to evaluate plasma and urinary levels of GDF-15 after pediatric renal transplantation (Rtx) and in children with chronic kidney disease (CKD) and its associations to cardiovascular risk factors. In this cross-sectional study, GDF-15 was measured in plasma and urine from 53 children with a renal transplant and 83 children with CKD and related to cardiovascular risk factors (hypertension, obesity, and cholesterol) and kidney function. Forty healthy children served as a control group. Plasma levels of GDF-15 (median and range) for a Tx (transplantation) cohort, CKD cohort, and healthy controls were, respectively, 865 ng/L (463-3039 ng/L), 508 ng/L (183-3279 ng/L), and 390 ng/L (306-657 ng/L). The CKD and Tx cohorts both had significantly higher GDF-15 levels than the control group (p < 0.001). Univariate associations between GDF-15 and hyperuricemia (p < 0.001), elevated triglycerides (p = 0.028), low HDL (p = 0.038), and obesity (p = 0.028) were found. However, mGFR (p < 0.001) and hemoglobin (p < 0.001) were the only significant predictors of GDF-15 in an adjusted analysis. Urinary GDF-15/creatinine ratios were 448 ng/mmol (74-5013 ng/mmol) and 540 ng/mmol (5-14960 ng/mmol) in the Tx cohort and CKD cohort, respectively. In the CKD cohort, it was weakly correlated to mGFR (r = -0.343, p = 0.002). Plasma levels of GDF-15 are elevated in children with CKD and after Rtx. The levels were not associated with traditional cardiovascular risk factors but strongly associated with renal function.
Collapse
Affiliation(s)
- Hjordis Thorsteinsdottir
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Norway
| | - Cathrin Lytomt Salvador
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Geir Mjøen
- Department of Nephrology, Oslo University Hospital, Norway
| | - Anine Lie
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Meryam Sugulle
- Division of Gynaecology and Obstetrics, Oslo University Hospital, Norway
| | - Camilla Tøndel
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Norway
| | - Atle Brun
- Laboratory for Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Norway
| | - Runar Almaas
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Norway
- Department of Pediatric Research, Oslo University Hospital, Norway
| | - Anna Bjerre
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
144
|
Carlsson AC, Nowak C, Lind L, Östgren CJ, Nyström FH, Sundström J, Carrero JJ, Riserus U, Ingelsson E, Fall T, Ärnlöv J. Growth differentiation factor 15 (GDF-15) is a potential biomarker of both diabetic kidney disease and future cardiovascular events in cohorts of individuals with type 2 diabetes: a proteomics approach. Ups J Med Sci 2020; 125:37-43. [PMID: 31805809 PMCID: PMC7054929 DOI: 10.1080/03009734.2019.1696430] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Diabetic kidney disease (DKD) is a leading risk factor for end-stage renal disease and is one of the most important risk factors for cardiovascular disease in patients with diabetes. It is possible that novel markers portraying the pathophysiological underpinning processes may be useful.Aim: To investigate the associations between 80 circulating proteins, measured by a proximity extension assay, and prevalent DKD and major adverse cardiovascular events (MACE) in type 2 diabetes.Methods: We randomly divided individuals with type 2 diabetes from three cohorts into a two-thirds discovery and one-third replication set (total n = 813, of whom 231 had DKD defined by estimated glomerular filtration rate <60 mg/mL/1.73 m2 and/or urinary albumin-creatinine ratio ≥3 g/mol). Proteins associated with DKD were also assessed as predictors for incident major adverse cardiovascular events (MACE) in persons with DKD at baseline.Results: Four proteins were positively associated with DKD in models adjusted for age, sex, cardiovascular risk factors, glucose control, and diabetes medication: kidney injury molecule-1 (KIM-1, odds ratio [OR] per standard deviation increment, 1.65, 95% confidence interval [CI] 1.27-2.14); growth differentiation factor 15 (GDF-15, OR 1.40, 95% CI 1.16-1.69); myoglobin (OR 1.57, 95% CI 1.30-1.91), and matrix metalloproteinase 10 (MMP-10, OR 1.43, 95% CI 1.17-1.74). In patients with DKD, GDF-15 was significantly associated with increased risk of MACE after adjustments for baseline age, sex, microalbuminuria, and kidney function and (59 MACE events during 7 years follow-up, hazard ratio per standard deviation increase 1.43 [95% CI 1.03-1.98]) but not after further adjustments for cardiovascular risk factors.Conclusion: Our proteomics approach confirms and extends previous associations of higher circulating levels of GDF-15 with both micro- and macrovascular disease in patients with type 2 diabetes. Our data encourage additional studies evaluating the clinical utility of our findings.
Collapse
Affiliation(s)
- Axel C. Carlsson
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
- CONTACT Axel C. Carlsson Department of Neurobiology Care Sciences and Society, Division for Family Medicine and Primary Care, 141 83 Huddinge, Sweden
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Johan Östgren
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Fredrik H. Nyström
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Juan Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Riserus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| |
Collapse
|
145
|
Egerstedt A, Berntsson J, Smith ML, Gidlöf O, Nilsson R, Benson M, Wells QS, Celik S, Lejonberg C, Farrell L, Sinha S, Shen D, Lundgren J, Rådegran G, Ngo D, Engström G, Yang Q, Wang TJ, Gerszten RE, Smith JG. Profiling of the plasma proteome across different stages of human heart failure. Nat Commun 2019; 10:5830. [PMID: 31862877 PMCID: PMC6925199 DOI: 10.1038/s41467-019-13306-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a major public health problem characterized by inability of the heart to maintain sufficient output of blood. The systematic characterization of circulating proteins across different stages of HF may provide pathophysiological insights and identify therapeutic targets. Here we report application of aptamer-based proteomics to identify proteins associated with prospective HF incidence in a population-based cohort, implicating modulation of immunological, complement, coagulation, natriuretic and matrix remodeling pathways up to two decades prior to overt disease onset. We observe further divergence of these proteins from the general population in advanced HF, and regression after heart transplantation. By leveraging coronary sinus samples and transcriptomic tools, we describe likely cardiac and specific cellular origins for several of the proteins, including Nt-proBNP, thrombospondin-2, interleukin-18 receptor, gelsolin, and activated C5. Our findings provide a broad perspective on both cardiac and systemic factors associated with HF development.
Collapse
Affiliation(s)
- Anna Egerstedt
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - John Berntsson
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Cardiovascular Epidemiology, Clinical Sciences, Lund University, Malmö, Sweden
| | - Maya Landenhed Smith
- Department of Cardiothoracic Surgery, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Olof Gidlöf
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Roland Nilsson
- Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Mark Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Quinn S Wells
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Selvi Celik
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Carl Lejonberg
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Sumita Sinha
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Dongxiao Shen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jakob Lundgren
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Department of Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden
| | - Göran Rådegran
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Department of Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden
| | - Debby Ngo
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Gunnar Engström
- Cardiovascular Epidemiology, Clinical Sciences, Lund University, Malmö, Sweden
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.
- Department of Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden.
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA.
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden.
| |
Collapse
|
146
|
Abstract
Objective: Despite several improvements in the management of heart failure (HF), it is still an incurable and a progressive disease. Several trials demonstrated that the process of inflammation may be responsible for initiation and progression of HF. The aim of the present study was to investigate the role of interleukin-33 (IL-33) in the pathogenesis of HF and to assess whether disease etiology and course of the disease affect the expression of cytokines. Methods: The study included 155 (106 male and 49 female) patients with systolic HF with a mean left ventricle ejection fraction of 32.13±12.8% and 60 (36 male and 24 female) healthy individuals. IL-33 concentrations were evaluated using enzyme-linked immunosorbent assay. Results: The concentration of IL-33 was statistically significantly lower in patients with HF than in healthy subjects, 16.91 (0–81.00) pg/mL and 92.51 (33.61–439.61) pg/mL, respectively. Patients with HF with ischemic etiology had lower concentration of IL-33 (10.75 pg/mL) than subjects with HF with non-ischemic etiology (21.05 pg/mL). Patients with stable HF (10.46 pg/mL) had lower IL-33 levels than those with unstable HF (19.02 pg/mL). Conclusion: The concentrations of IL-33 were lower in patients with HF than in healthy controls, which may play an important role of above cytokine in HF development and progression. In addition, interleukin concentrations varied depending on the etiology and severity of the course of the disease.
Collapse
|
147
|
ST2 levels increased and were associated with changes in left ventricular systolic function during a three-year follow-up after adjuvant radiotherapy for breast cancer. Breast 2019; 49:183-186. [PMID: 31862685 PMCID: PMC7375584 DOI: 10.1016/j.breast.2019.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/04/2019] [Accepted: 12/01/2019] [Indexed: 01/30/2023] Open
Abstract
Objectives To search for biomarkers of RT-induced cardiotoxicity, we studied the behavior of ST2 during RT and three years after RT, and the associations with echocardiographic changes. Materials and methods We measured soluble ST2 (ng/ml) in serum samples from 63 patients receiving RT for early breast cancer. Sampling and echocardiography were performed at baseline, after RT and at the three-year follow-up. Patients were grouped by >15% (group 1) and ≤15% (group 2) relative worsening in global longitudinal strain (GLS). Results ST2 levels tended to increase during RT, from a median (interquartile range; IQR) of 17.9 (12.4–22.4) at baseline to 18.2 (14.1–23.5) after RT (p = 0.075). By the three-year follow up, ST2 levels increased to 18.7 (15.8–24.2), p = 0.018. The increase in ST2 level was associated with worsening cardiac systolic function at three-year follow-up, GLS (rho = 0.272, p = 0.034) and left ventricular ejection fraction (LVEF) (rho = ─0.343, p = 0.006). Group 1 (n = 14) had a significant increase in ST2 levels from 17.8 (12.3–22.5) at baseline to 18.4 (15.6–22.6) after RT, p = 0.035 and to 19.9 (16.0–25.1) three years after RT, p = 0.005. ST2 levels were stable in group 2 (n = 47): 17.8 (12.3–22.0) at baseline, 17.7 (12.6–23.5) after RT and 18.0 (15.5–22.4) at three years. Conclusion ST2 may be useful for determining which patients are at risk for long-term cardiovascular toxicity following adjuvant breast cancer RT, but prospective clinical studies are needed to confirm this hypothesis. ST2 levels increase 3 years from adjuvant radiotherapy for breast cancer. The increase in ST2 levels is associated with a worsening in global longitudinal strain (GLS). Increase in ST2 levels is seen during radiotherapy in patients with a >15% change in GLS over 3 years.
Collapse
|
148
|
Bansal N, Zelnick L, Shlipak MG, Anderson A, Christenson R, Deo R, deFilippi C, Feldman H, Lash J, He J, Kusek J, Ky B, Seliger S, Soliman EZ, Go AS. Cardiac and Stress Biomarkers and Chronic Kidney Disease Progression: The CRIC Study. Clin Chem 2019; 65:1448-1457. [PMID: 31578216 PMCID: PMC6927328 DOI: 10.1373/clinchem.2019.305797] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/27/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Increases in cardiac and stress biomarkers may be associated with loss of kidney function through shared mechanisms involving cardiac and kidney injury. We evaluated the associations of cardiac and stress biomarkers [N-terminal pro-B-type natriuretic peptide (NT-proBNP), high-sensitivity troponin T (hsTnT), growth differentiation factor 15 (GDF-15), soluble ST-2 (sST-2)] with progression of chronic kidney disease (CKD). METHODS We included 3664 participants with CKD from the Chronic Renal Insufficiency Cohort study. All biomarkers were measured at entry. The primary outcome was CKD progression, defined as progression to end-stage renal disease (ESRD) or 50% decline in estimated glomerular filtration rate (eGFR). Cox models tested the association of each biomarker with CKD progression, adjusting for demographics, site, diabetes, cardiovascular disease, eGFR, urine proteinuria, blood pressure, body mass index, cholesterol, medication use, and mineral metabolism. RESULTS There were 1221 participants who had CKD progression over a median (interquartile range) follow-up of 5.8 (2.4-8.6) years. GDF-15, but not sST2, was significantly associated with an increased risk of CKD progression [hazard ratios (HRs) are per SD increase in log-transformed biomarker]: GDF-15 (HR, 1.50; 95% CI, 1.35-1.67) and sST2 (HR, 1.07; 95% CI, 0.99-1.14). NT-proBNP and hsTnT were also associated with increased risk of CKD progression, but weaker than GDF-15: NT-proBNP (HR, 1.24; 95% CI, 1.13-1.36) and hsTnT (HR, 1.11; 95% CI, 1.01-1.22). CONCLUSIONS Increases in GDF-15, NT-proBNP, and hsTnT are associated with greater risk for CKD progression. These biomarkers may inform mechanisms underlying kidney injury.
Collapse
Affiliation(s)
- Nisha Bansal
- Department of Medicine, University of Washington, Seattle, WA;
| | - Leila Zelnick
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Amanda Anderson
- Department of Epidemiology, Tulane University, New Orleans, LA
| | | | - Rajat Deo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Harold Feldman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - James Lash
- Department of Medicine, University of Illinois, Chicago, IL
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA
| | - John Kusek
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bonnie Ky
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephen Seliger
- Department of Laboratory Science, University of Maryland, Baltimore, MD
| | | | - Alan S Go
- Division of Research, Kaiser Permanente Northern California Division of Research, Oakland, CA
| |
Collapse
|
149
|
Arkoumani M, Papadopoulou-Marketou N, Nicolaides NC, Kanaka-Gantenbein C, Tentolouris N, Papassotiriou I. The clinical impact of growth differentiation factor-15 in heart disease: A 2019 update. Crit Rev Clin Lab Sci 2019; 57:114-125. [PMID: 31663791 DOI: 10.1080/10408363.2019.1678565] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Growth differentiation factor-15 (GDF-15), also known as macrophage inhibitory cytokine-1 (MIC-1) or non-steroidal anti-inflammatory drug-activated gene (NAG-1) has been identified as a biomarker of response to treatment and prognosis in cardiovascular diseases. GDF-15 is a member of the transforming growth factor-β superfamily and is involved in several pathological conditions such as inflammation, cancer, cardiovascular, pulmonary and renal diseases. Cardiac myocytes produce and secrete GDF-15 in response to oxidative stress, stimulation with angiotensin II or proinflammatory cytokines, ischemia, and mechanical stretch. Other cellular sources of GDF-15 production are macrophages, vascular smooth muscle cells, endothelial cells, and adipocytes, which secrete GDF-15 in response to oxidative or metabolic stress or stimulation of proinflammatory cytokines. GDF-15 is induced in hypertrophic and dilated cardiomyopathy after volume overload, ischemia, and heart failure. GDF-15 can be used as a marker of prognosis in patients with cardiovascular disorders, in combination with conventional prognostic factors, such as N-terminal pro B-type natriuretic peptide (NT-proBNP) and high-sensitivity troponin T (hs-TnT).
Collapse
Affiliation(s)
- Maria Arkoumani
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece.,First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Nektaria Papadopoulou-Marketou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece.,First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Nicolas C Nicolaides
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Christina Kanaka-Gantenbein
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
150
|
Bansal N, Zelnick L, Go A, Anderson A, Christenson R, Deo R, Defilippi C, Lash J, He J, Ky B, Seliger S, Soliman E, Shlipak M. Cardiac Biomarkers and Risk of Incident Heart Failure in Chronic Kidney Disease: The CRIC (Chronic Renal Insufficiency Cohort) Study. J Am Heart Assoc 2019; 8:e012336. [PMID: 31645163 PMCID: PMC6898812 DOI: 10.1161/jaha.119.012336] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Cardiac biomarkers may signal mechanistic pathways involved in heart failure (HF), a leading complication in chronic kidney disease. We tested the associations of NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide), high‐sensitivity troponin T (hsTnT), galectin‐3, growth differentiation factor‐15 (GDF‐15), and soluble ST2 (sST2) with incident HF in chronic kidney disease. Methods and Results We examined adults with chronic kidney disease enrolled in a prospective, multicenter study. All biomarkers were measured at baseline. The primary outcome was incident HF. Secondary outcomes included HF with preserved ejection fraction (EF≥50%) and reduced ejection fraction (EF<50%). Cox models were used to test the association of each cardiac biomarker with HF, adjusting for demographics, kidney function, cardiovascular risk factors, and medication use. Among 3314 participants, all biomarkers, with the exception of galectin‐3, were significantly associated with increased risk of incident HF (hazard ratio per SD higher concentration of log‐transformed biomarker): NT‐proBNP (hazard ratio, 2.07; 95% CI, 1.79–2.39); hsTnT (hazard ratio, 1.38; 95% CI, 1.21–1.56); GDF‐15 (hazard ratio, 1.44; 95% CI, 1.26–1.66) and sST2 (hazard ratio, 1.19; 95% CI, 1.05–1.35). Higher NT‐proBNP, hsTnT, and GDF‐15 were also associated with a greater risk of HF with reduced EF; while higher NT‐proBNP GDF‐15 and sST2 were associated with HF with preserved EF. Galectin‐3 was not associated with either HF with reduced EF or HF with preserved EF. Conclusions In chronic kidney disease, elevations of NT‐proBNP, hsTnT, GDF‐15, sST2 were associated with incident HF. There was a borderline association of galectin‐3 with incident HF. NT‐proBNP and hsTnT were more strongly associated with HF with reduced EF, while the associations of the newer biomarkers GDF‐15 and sST2 were stronger for HF with preserved EF.
Collapse
Affiliation(s)
| | | | - Alan Go
- Division of Research Kaiser Permanente Northern California Oakland CA
| | | | | | - Rajat Deo
- University of Pennsylvania Philadelphia PA
| | | | | | - Jiang He
- Tulane University New Orleans LA
| | - Bonnie Ky
- University of Pennsylvania Philadelphia PA
| | | | | | | | | | | |
Collapse
|