1
|
Duus Holle SL, Søholm H, Møller JE, Winther-Jensen M, Kunkel JB, Okkels Jensen L, Kjærgaard J, Lindholm MG, Helgestad OKL, Wiberg S, Sousa RR, Holmvang L, Thomsen JH, Goetze JP, Hassager C, Frydland M. The prognostic value of neurohormonal and inflammatory biomarkers in addition to the TIMI risk score in patients with ST-elevation myocardial infarction. Biomarkers 2025; 30:1-9. [PMID: 39773138 DOI: 10.1080/1354750x.2024.2435866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The Thrombolysis in Myocardial Infarction (TIMI) risk score estimates mortality for patients with ST-elevation myocardial infarction (STEMI). This study aimed to investigate whether biomarkers reflecting the neurohormonal response (pro-atrial natriuretic peptide (proANP), mid-regional pro-adrenomedullin (MR-proADM), and copeptin), inflammation (suppression of tumorigenicity 2 (ST2), C-reactive protein (CRP), and leukocytes), and troponin add prognostic value to the TIMI risk score. METHODS This sub-study of the prospective PREDICT cohort included 1700 non-comatose and non-cardiogenic shock STEMI patients upon admission. Blood samples were collected before coronary angiography. Biomarker quartiles (Q4vsQ1-3) association with 30-day mortality were examined using Cox proportional hazard models. RESULTS High levels of all biomarkers were associated with 30-day mortality independently of TIMI risk score, hazard ratio (HR)Q4vsQ1-3 (95%CI), MR-proADM: 8.8 (3.9-20), proANP: 3.5 (1.8-6.7), copeptin: 1.9 (1.1-3.5), ST2: 4.5 (2.3-8.6), CRP: 2.6 (1.3-4.9), and leukocyte: 2.18 (1.2;4.0). TIMI risk score had a high prognostic value, AUC(95%CI): 0.76 (0.69-0.83). Only MR-proADM, proANP, CRP, ST2, and TnT added prognostic value to the risk score, 0.84 (0.77-0.91), 0.80 (0.74-0.87), 0.78 (0.71-0.86), 0.81 (0.73-0.88), and 0.79 (0.71-0.87), respectively. However, MR-proADM demonstrated a higher prognostic value on its own (0.86 (0.80-0.91)). CONCLUSION TIMI risk score and all the biomarkers added prognostic values of 30-day mortality. The strongest predictor of 30-day mortality was observed for MR-proADM alone.
Collapse
Affiliation(s)
- Sarah Louise Duus Holle
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Helle Søholm
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Zealand University Hospital, Roskilde, Denmark
| | | | - Matilde Winther-Jensen
- Section of Data, Biostatistics and Pharmacoepidemiology, Center for Clinical Research and Prevention, Frederiksberg Hospital, Frederiksberg, Denmark
| | - Joakim Bo Kunkel
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Jesper Kjærgaard
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Sebastian Wiberg
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiothoracic Anaesthesiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Lene Holmvang
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jakob Hartvig Thomsen
- Department of Cardiology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Jens Peter Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- The Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Christian Hassager
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- The Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Martin Frydland
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Huang S, Yu LJ, Sun GF, Zhang ZX. Short-term predictive value of sST2 in patients with STEMI following primary PCI: a prospective observational study. BMC Cardiovasc Disord 2025; 25:21. [PMID: 39819309 PMCID: PMC11737151 DOI: 10.1186/s12872-025-04488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
AIM The objective of this study was to investigate the level of soluble suppression of tumorigenicity-2 (sST2) in patients with acute ST-segment elevation myocardial infarction (STEMI) following primary percutaneous coronary intervention (PCI), and to provide a new biomarker for clinical management and prognosis assessment. METHOD This was a prospective study. 148 STEMI patients following primary PCI were enrolled and divided into 2 groups by the median value of sST2 and afterwards followed up for 30 days to access the occurrence of major adverse cardiac events (MACEs), which were defined as cardiovascular death, heart failure and recurrent MI. RESULTS sST2 ranged from 20.57 to 98.96 ng/mL. High sST2 group had higher MACEs rate compared to low sST2 group (28.8% vs. 8.0%, P = 0.001). sST2 was positively correlated with age (r = 0.181, P = 0.027), SYNTAX score (r = 0.257, P = 0.002), high-sensitive C-reactive protein (hs-CRP) (r = 0.225, P = 0.006), B-type natriuretic peptide (BNP) (r = 0.225, P = 0.006) and negatively with left ventricle ejection fraction (LVEF) (r = -0.197, P = 0.016). After adjustment for clinical variables, sST2 level (OR 3.680, P = 0.015) and LVEF (OR 0.880, P < 0.001) remained independent predictors of 30-days MACEs. In receiver operating characteristic curve (ROC) analyses, the area under the curve (AUC) of sST2 for predicting 30-days MACEs was 0.755(P < 0.001). The AUC of sST2 combining hs-CRP and LVEF for prediction was 0.828(95%CI [0.743-0.912], P < 0.001). CONCLUSION sST2 level after primary PCI was an independent risk factor of MACEs in STEMI patients through 30 days follow-up.
Collapse
Affiliation(s)
- Shan Huang
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Huli District, Xiamen, China
| | - Lu-Jiao Yu
- Department of Geriatrics, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China
| | - Guang-Feng Sun
- Department of Emergency, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Xiamen, China
| | - Zi-Xin Zhang
- Department of Cardiology, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China.
| |
Collapse
|
3
|
Zhang W, Zhang J, Wang Z, Li T, Liu C, Kang X, Cui X, Yang J, Qu H, Duanmu J, Peng Y, Wang K, Jin L, Xie P, Zheng W, Shang H, Liu Y, Tian Z, Liu Z, Jin Y, Li Y, Li N, Zhuo X, Wu Y, Shi X, Ma R, Sun Y, Zhang K, Fang X, Hu X, Dong E, Zhang S, Zhang Y. Extracellular RIPK3 Acts as a Damage-Associated Molecular Pattern to Exaggerate Cardiac Ischemia/Reperfusion Injury. Circulation 2024; 150:1791-1811. [PMID: 39411860 DOI: 10.1161/circulationaha.123.068595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/05/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Cardiac ischemia/reperfusion (I/R) injury has emerged as an important therapeutic target for ischemic heart disease. Currently, there is no effective therapy for reducing cardiac I/R injury. Damage-associated molecular patterns are endogenous molecules released after cellular damage to exaggerate tissue inflammation and injury. RIPK3 (receptor-interacting protein kinase 3), a well-established intracellular mediator of cell necroptosis and inflammation, serves as a circulating biomarker of multiple diseases. However, whether extracellular RIPK3 also exerts biological functions in cardiac I/R injury remains totally unknown. METHODS Patients with acute myocardial infarction receiving percutaneous coronary intervention (PCI) were recruited independently in the discovery cohort (103 patients) and validation cohort (334 patients), and major adverse cardiovascular events were recorded. Plasma samples were collected before and after PCI (6 and 24 h) for RIPK3 concentration measurement. Cultured neonatal rat ventricular myocytes, macrophages and endothelial cells, and in vivo mouse models with myocardial injury induced by I/R (or hypoxia/reoxygenation) were used to investigate the role and mechanisms of extracellular RIPK3. Another cohort including patients with acute myocardial infarction receiving PCI and healthy volunteers was recruited to further explore the mechanisms of extracellular RIPK3. RESULTS In the discovery cohort, elevated plasma RIPK3 levels after PCI are associated with poorer short- and long-term outcomes in patients with acute myocardial infarction, as confirmed in the validation cohort. In both cultured cells and in vivo mouse models, recombinant RIPK3 protein exaggerated myocardial I/R (or hypoxia/reoxygenation) injury, which was alleviated by the RIPK3 antibody. Mechanistically, RIPK3 acted as a damage-associated molecular pattern and bound with RAGE (receptor of advanced glycation end-products), subsequently activating CaMKII (Ca2+/calmodulin-dependent kinase II) to elicit the detrimental effects. The positive correlation between plasma RIPK3 concentrations and CaMKII phosphorylation in human peripheral blood mononuclear cells was confirmed. CONCLUSIONS We identified the positive relationship between plasma RIPK3 concentrations and the risk of major adverse cardiovascular events in patients with acute myocardial infarction receiving PCI. As a damage-associated molecular pattern, extracellular RIPK3 plays a causal role in multiple pathological conditions during cardiac I/R injury through RAGE/CaMKII signaling. These findings expand our understanding of the physiological and pathological roles of RIPK3, and also provide a promising therapeutic target for myocardial I/R injury and the associated complications.
Collapse
Affiliation(s)
- Wenjia Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Junxia Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing (J.Z., E.D.)
| | - Zeyuan Wang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Ting Li
- Department of Cardiology, First Affiliated Hospital, Xi'an Jiaotong University, China (T.L., X.Z., Y.W.)
| | - Changyun Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Xiaomeng Cui
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Jingli Yang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Huilin Qu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Jiaxin Duanmu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Ying Peng
- Department of General Surgery (Y.P), Peking University Third Hospital, Beijing, China
| | - Kai Wang
- Department of Hysiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Female Fertility Promotion, Center for Reproductive III Medicine (K.W.), Peking University, Beijing, China
| | - Li Jin
- State Key Laboratory of Membrane Biology Institute of Molecular Medicine, College of Future Technology (L.J., P.X., W. Zheng, H.S., X.H.), Peking University, Beijing, China
| | - Peng Xie
- State Key Laboratory of Membrane Biology Institute of Molecular Medicine, College of Future Technology (L.J., P.X., W. Zheng, H.S., X.H.), Peking University, Beijing, China
| | - Wen Zheng
- State Key Laboratory of Membrane Biology Institute of Molecular Medicine, College of Future Technology (L.J., P.X., W. Zheng, H.S., X.H.), Peking University, Beijing, China
| | - Haibao Shang
- State Key Laboratory of Membrane Biology Institute of Molecular Medicine, College of Future Technology (L.J., P.X., W. Zheng, H.S., X.H.), Peking University, Beijing, China
| | - Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Zhuang Tian
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Zhenyu Liu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Ye Jin
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China (Y.J.)
| | - Yingjia Li
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
| | - Nan Li
- Research Center of Clinical Epidemiology (N.L.), Peking University Third Hospital, Beijing, China
| | - Xiaozhen Zhuo
- Department of Cardiology, First Affiliated Hospital, Xi'an Jiaotong University, China (T.L., X.Z., Y.W.)
| | - Yue Wu
- Department of Cardiology, First Affiliated Hospital, Xi'an Jiaotong University, China (T.L., X.Z., Y.W.)
| | - Xiaolu Shi
- State Key Laboratory of Membrane Biology Institute of Molecular Medicine, College of Future Technology (L.J., P.X., W. Zheng, H.S., X.H.), Peking University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences (X.S., R.M.)
| | - Runhao Ma
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences (X.S., R.M.)
| | - Yueshen Sun
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Kai Zhang
- Department of Anesthesiology and Intensive Care, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (K.Z., X.F.)
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (K.Z., X.F.)
| | - Xiaomin Hu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing (J.Z., E.D.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, China (E.D.)
| | - Shuyang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing (Z.W., Z.T., Z.L., Y.J., Y.S., X.H., S.Z.)
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center (W. Zhang, J.Z., C.L., X.K., X.C., J.Y, H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z)
- Department of Cardiology and Institute of Vascular Medicine (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D., Y. Liu, Y. Li, E.D., Y.Z.)
- Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling (W. Zhang, J.Z., C.L., X.K., X.C., J.Y., H.Q., J.D, Y. Liu, Y. Li, E.D., Y.Z.)
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (Y.Z.)
| |
Collapse
|
4
|
Yadigaroğlu M, Güzel M, Erdem E, Görgün S, Aksu EA, Ocak M, Yadigaroğlu NÖ, Demir MT, Yücel M. IL-33/sST2 signaling pathway in pulmonary thromboembolism: A clinical observational study. Cytokine 2024; 182:156707. [PMID: 39084069 DOI: 10.1016/j.cyto.2024.156707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Pulmonary thromboembolism (PTE) is a cardiovascular emergency that can result in mortality. In the interleukin-33 (IL-33) /soluble suppression of tumorigenicity 2 (sST2) signaling pathway, increased sST2 is a cardiovascular risk factor. This study aimed to investigate the effectiveness of biomarkers in the IL-33/sST2 signaling pathway in determining PTE diagnosis, clinical severity, and mortality. METHOD This study was conducted as a single-center, prospective, observational study. Patients admitted to the emergency department and diagnosed with PTE constituted the patient group (n = 112), and healthy volunteers with similar sociodemographic characteristics constituted the control group (n = 62). Biomarkers in the IL-33/sST2 signaling pathway were evaluated for diagnosis, clinical severity, and prognosis. RESULTS IL-33 was lower in the patient group than in the control group (275.89 versus 403.35 pg/mL), while sST2 levels were higher in the patient group than in the control group (53.16 versus 11.78 ng/mL) (p < 0.001 and p = 0.001; respectively). The AUC of IL-33 to diagnose PTE was 0.656 (95 % CI: 0.580-0.726). The optimal IL-33 cut-off point to diagnose PTE was ≤304.11 pg/mL (56.2 % sensitivity, 79 % specificity). The AUC of sST2 to diagnose PTE was 0.818 (95 % CI: 0.752-0.872). The optimal sST2 cut-off point to diagnose PTE was >14.48 ng/mL (83 % sensitivity, 71 % specificity). IL-33 levels were lower in patients with mortality (169.85 versus 332.04 pg/mL) compared to patients without mortality, whereas sST2 levels were higher in patients with mortality (118.32 versus 28.07 ng/mL) compared to patients without mortality (p > 0.001 for both). The AUC of IL-33 to predict the mortality of PTE was 0.801 (95 % CI: 0.715-0.870). The optimal IL-33 cut-off point to predict the mortality of PTE was ≤212.05 pg/mL (75 % sensitivity, 79.5 % specificity). The AUC of sST2 to predict the mortality of PTE was 0.824 (95 % CI: 0.740-0.889). The optimal sST2 cut-off point to predict the mortality of PTE was >81 ng/mL (95.8 % sensitivity, 78.4 % specificity). CONCLUSION In the IL-33/ST2 signaling pathway, decreased IL-33 and increased sST2 are valuable biomarkers for diagnosis and prediction of mortality in patients with PTE.
Collapse
Affiliation(s)
- Metin Yadigaroğlu
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | - Murat Güzel
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | - Elif Erdem
- Şırnak State Hospital, Department of Emergency Medicine, Şırnak, Turkey.
| | - Selim Görgün
- Samsun Education and Research Hospital, Department of Microbiology and Clinical Microbiology, Samsun, Turkey.
| | - Esra Arslan Aksu
- Samsun University, Faculty of Medicine, Department of Chest Diseases, Samsun, Turkey.
| | - Metin Ocak
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | | | - Mehmet Tevfik Demir
- Samsun Education and Research Hospital, Department of Emergency Medicine, Samsun, Turkey.
| | - Murat Yücel
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| |
Collapse
|
5
|
Feng Y, He LQ. Soluble ST2: A Novel Biomarker for Diagnosis and Prognosis of Cardiovascular Disease. Curr Med Sci 2024; 44:669-679. [PMID: 39096477 DOI: 10.1007/s11596-024-2907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
The increasing incidence of cardiovascular disease (CVD) is a significant global health concern, affecting millions of individuals each year. Accurate diagnosis of acute CVD poses a formidable challenge, as misdiagnosis can significantly decrease patient survival rates. Traditional biomarkers have played a vital role in the diagnosis and prognosis of CVDs, but they can be influenced by various factors, such as age, sex, and renal function. Soluble ST2 (sST2) is a novel biomarker that is closely associated with different CVDs. Its low reference change value makes it suitable for continuous measurement, unaffected by age, kidney function, and other confounding factors, facilitating risk stratification of CVDs. Furthermore, the combination of sST2 with other biomarkers can enhance diagnostic accuracy and prognostic value. This review aims to provide a comprehensive overview of sST2, focusing on its diagnostic and prognostic value as a myocardial marker for different types of CVDs and discussing the current limitations of sST2.
Collapse
Affiliation(s)
- Yin Feng
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Qun He
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
van Blokland IV, Oelen R, Groot HE, Benjamins JW, Pekayvaz K, Losert C, Knottenberg V, Heinig M, Nicolai L, Stark K, van der Harst P, Franke L, van der Wijst MG. Single-Cell Dissection of the Immune Response After Acute Myocardial Infarction. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004374. [PMID: 38752343 PMCID: PMC11188632 DOI: 10.1161/circgen.123.004374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/17/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The immune system's role in ST-segment-elevated myocardial infarction (STEMI) remains poorly characterized but is an important driver of recurrent cardiovascular events. While anti-inflammatory drugs show promise in reducing recurrence risk, their broad immune system impairment may induce severe side effects. To overcome these challenges, a nuanced understanding of the immune response to STEMI is needed. METHODS For this, we compared peripheral blood mononuclear single-cell RNA-sequencing (scRNA-seq) and plasma protein expression over time (hospital admission, 24 hours, and 6-8 weeks post-STEMI) in 38 patients and 38 controls (95 995 diseased and 33 878 control peripheral blood mononuclear cells). RESULTS Compared with controls, classical monocytes were increased and CD56dim natural killer cells were decreased in patients with STEMI at admission and persisted until 24 hours post-STEMI. The largest gene expression changes were observed in monocytes, associating with changes in toll-like receptor, interferon, and interleukin signaling activity. Finally, a targeted cardiovascular biomarker panel revealed expression changes in 33/92 plasma proteins post-STEMI. Interestingly, interleukin-6R, MMP9 (matrix metalloproteinase-9), and LDLR (low-density lipoprotein receptor) were affected by coronary artery disease-associated genetic risk variation, disease status, and time post-STEMI, indicating the importance of considering these aspects when defining potential future therapies. CONCLUSIONS Our analyses revealed the immunologic pathways disturbed by STEMI, specifying affected cell types and disease stages. Additionally, we provide insights into patients expected to benefit most from anti-inflammatory treatments by identifying the genetic variants and disease stage at which these variants affect the outcome of these (drug-targeted) pathways. These findings advance our knowledge of the immune response post-STEMI and provide guidance for future therapeutic studies.
Collapse
Affiliation(s)
- Irene V. van Blokland
- Department of Cardiology (I.V.B., H.E.G., J.W.B.), University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics (I.V.B., R.O., L.F., M.G.P.v.d.W.), University Medical Center Groningen, Groningen, the Netherlands
| | - Roy Oelen
- Department of Genetics (I.V.B., R.O., L.F., M.G.P.v.d.W.), University Medical Center Groningen, Groningen, the Netherlands
| | - Hilde E. Groot
- Department of Cardiology (I.V.B., H.E.G., J.W.B.), University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Walter Benjamins
- Department of Cardiology (I.V.B., H.E.G., J.W.B.), University Medical Center Groningen, Groningen, the Netherlands
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital, Ludwig-Maximilian University, Munich, Germany (K.P., V.K., L.N., K.S.)
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany (K.P., V.K., L.N., K.S.)
| | - Corinna Losert
- Institute of Computational Biology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (C.L., M.H.)
- Department of Computer Science, TUM School of Computation, Information & Technology, Garching, Germany (C.L., M.H.)
| | - Viktoria Knottenberg
- Medizinische Klinik und Poliklinik I, University Hospital, Ludwig-Maximilian University, Munich, Germany (K.P., V.K., L.N., K.S.)
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany (K.P., V.K., L.N., K.S.)
| | - Matthias Heinig
- Institute of Computational Biology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany (C.L., M.H.)
- Department of Computer Science, TUM School of Computation, Information & Technology, Garching, Germany (C.L., M.H.)
- Department of Informatics, Ludwig-Maximilians Universität München, Munich, Germany (M.H.)
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital, Ludwig-Maximilian University, Munich, Germany (K.P., V.K., L.N., K.S.)
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany (K.P., V.K., L.N., K.S.)
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital, Ludwig-Maximilian University, Munich, Germany (K.P., V.K., L.N., K.S.)
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany (K.P., V.K., L.N., K.S.)
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands (P.v.d.H.)
| | - Lude Franke
- Department of Genetics (I.V.B., R.O., L.F., M.G.P.v.d.W.), University Medical Center Groningen, Groningen, the Netherlands
| | - Monique G.P. van der Wijst
- Department of Genetics (I.V.B., R.O., L.F., M.G.P.v.d.W.), University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
7
|
Piccioni A, Baroni S, Manca F, Sarlo F, Savioli G, Candelli M, Bronzino A, Covino M, Gasbarrini A, Franceschi F. Multi-Marker Approach in Patients with Acute Chest Pain in the Emergency Department. J Pers Med 2024; 14:564. [PMID: 38929785 PMCID: PMC11204991 DOI: 10.3390/jpm14060564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Chest pain is a prevalent reason for emergency room referrals and presents diagnostic challenges. The physician must carefully differentiate between cardiac and noncardiac causes, including various vascular and extracardiovascular conditions. However, it is crucial not to overlook serious conditions such as acute coronary syndrome (ACS). Diagnosis of acute myocardial infarction (AMI) and early discharge management become difficult when traditional clinical criteria, ECG, and troponin values are insufficient. Recently, the focus has shifted to a "multi-marker" approach to improve diagnostic accuracy and prognosis in patients with chest pain. METHODS This observational, prospective, single-center study involved, with informed consent, 360 patients presenting to the emergency department with typical chest pain and included a control group of 120 healthy subjects. In addition to routine examinations, including tests for hsTnI (Siemens TNIH kit), according to the 0-1 h algorithm, biochemical markers sST2 (tumorigenicity suppression-2) and suPAR (soluble urokinase plasminogen activator receptor) were also evaluated for each patient. A 12-month follow-up was conducted to monitor outcomes and adverse events. RESULTS We identified two groups of patients: a positive one (112 patients) with high levels of hsTnI, sST2 > 24.19 ng/mL, and suPAR > 2.9 ng/mL, diagnosed with ACS; and a negative one (136 patients) with low levels of hsTnI, suPAR < 2.9 ng/mL, and sST2 < 24.19 ng/mL. During the 12-month follow-up, no adverse events were observed in the negative group. In the intermediate group, patients with hsTnI between 6 ng/L and the ischemic limit, sST2 > 29.1 ng/mL and suPAR > 2.9 ng/mL, showed the highest probability of adverse events during follow-up, while those with sST2 < 24.19 ng/mL and suPAR < 2.9 ng/mL had a better outcome with no adverse events at 12 months. CONCLUSION Our data suggest that sST2 and suPAR, together with hsTnI, may be useful in the prognosis of cardiovascular patients with ACS, providing additional information on endothelial damage. These biomarkers could guide the clinical decision on further diagnostic investigations. In addition, suPAR and sST2 emerge as promising for event prediction in patients with chest pain. Their integration into the standard approach in PS could facilitate more efficient patient management, allowing safe release or timely admission based on individual risk.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| | - Silvia Baroni
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (S.B.); (F.S.)
| | - Federica Manca
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| | - Francesca Sarlo
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (S.B.); (F.S.)
| | - Gabriele Savioli
- Departement of Emergency, IRCCS Fondazione Policlinico San Matteo, 27100 Pavia, Italy;
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| | - Alessandra Bronzino
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| | - Marcello Covino
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| | - Antonio Gasbarrini
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Roma, Italy; (F.M.); (M.C.); (A.B.); (M.C.); (F.F.)
| |
Collapse
|
8
|
Numan L, Aarts E, Ramjankhan F, Oerlemans MIF, van der Meer MG, de Jonge N, Oppelaar A, Kemperman H, Asselbergs FW, Van Laake LW. Soluble Suppression of Tumorigenicity-2 Predicts Mortality and Right Heart Failure in Patients With a Left Ventricular Assist Device. J Am Heart Assoc 2024; 13:e029827. [PMID: 38193339 PMCID: PMC10926819 DOI: 10.1161/jaha.123.029827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 11/01/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Soluble suppression of tumorigenicity-2 (sST2) predicts mortality in patients with heart failure. The predictive value of sST2 in patients with a left ventricular assist device remains unknown. Therefore, we studied the relationship between sST2 and outcome after left ventricular assist device implantation. METHODS AND RESULTS sST2 levels of patients with a left ventricular assist device implanted between January 2015 and December 2022 were included in this observational study. The median follow-up was 25 months, during which 1573 postoperative sST2 levels were measured in 199 patients, with a median of 29 ng/mL. Survival of patients with normal and elevated preoperative levels was compared using Kaplan-Meier analysis, which did not differ significantly (P=0.22) between both groups. The relationship between postoperative sST2, survival, and right heart failure was evaluated using a joint model, which showed a significant relationship between the absolute sST2 level and mortality, with a hazard ratio (HR) of 1.20 (95% CI, 1.10-1.130; P<0.01) and an HR of 1.22 (95% CI, 1.07-1.39; P=0.01) for right heart failure, both per 10-unit sST2 increase. The sST2 instantaneous change was not predictive for survival or right heart failure (P=0.99 and P=0.94, respectively). Multivariate joint model analysis showed a significant relationship between sST2 with mortality adjusted for NT-proBNP (N-terminal pro-B-type natriuretic peptide), with an HR of 1.19 (95% CI, 1.00-1.42; P=0.05), whereas the HR of right heart failure was not significant (1.22 [95% CI, 0.94-1.59]; P=0.14), both per 10-unit sST2 increase. CONCLUSIONS Time-dependent postoperative sST2 predicts all-cause mortality after left ventricular assist device implantation after adjustment for NT-proBNP. Future research is warranted into possible target interventions and the optimal monitoring frequency.
Collapse
Affiliation(s)
- Lieke Numan
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
| | - Emmeke Aarts
- Department of Methodology and StatisticsUtrecht UniversityUtrechtthe Netherlands
| | - Faiz Ramjankhan
- Department of Cardiothoracic SurgeryUniversity Medical Center Utrecht, University of UtrechtUtrechtthe Netherlands
| | - Marish I. F. Oerlemans
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
| | - Manon G. van der Meer
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
| | - Nicolaas de Jonge
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
| | - Anne‐Marie Oppelaar
- Department of Cardiothoracic SurgeryUniversity Medical Center Utrecht, University of UtrechtUtrechtthe Netherlands
| | - Hans Kemperman
- Department of Central Diagnostic LaboratoryUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Folkert W. Asselbergs
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health SciencesUniversity College LondonLondonUnited Kingdom
- Health Data Research UK and Institute of Health InformaticsUniversity College LondonLondonUnited Kingdom
- Department of Cardiology, Amsterdam Cardiovascular SciencesAmsterdam University Medical Centre, University of AmsterdamAmsterdamthe Netherlands
| | - Linda W. Van Laake
- Department of CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtthe Netherlands
| |
Collapse
|
9
|
Chatterton C, Romero R, Jung E, Gallo DM, Suksai M, Diaz-Primera R, Erez O, Chaemsaithong P, Tarca AL, Gotsch F, Bosco M, Chaiworapongsa T. A biomarker for bacteremia in pregnant women with acute pyelonephritis: soluble suppressor of tumorigenicity 2 or sST2. J Matern Fetal Neonatal Med 2023; 36:2183470. [PMID: 36997168 PMCID: PMC10352993 DOI: 10.1080/14767058.2023.2183470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 02/15/2023] [Indexed: 04/01/2023]
Abstract
Objective: Sepsis is a leading cause of maternal death, and its diagnosis during the golden hour is critical to improve survival. Acute pyelonephritis in pregnancy is a risk factor for obstetrical and medical complications, and it is a major cause of sepsis, as bacteremia complicates 15-20% of pyelonephritis episodes in pregnancy. The diagnosis of bacteremia currently relies on blood cultures, whereas a rapid test could allow timely management and improved outcomes. Soluble suppression of tumorigenicity 2 (sST2) was previously proposed as a biomarker for sepsis in non-pregnant adults and children. This study was designed to determine whether maternal plasma concentrations of sST2 in pregnant patients with pyelonephritis can help to identify those at risk for bacteremia.Study design: This cross-sectional study included women with normal pregnancy (n = 131) and pregnant women with acute pyelonephritis (n = 36). Acute pyelonephritis was diagnosed based on a combination of clinical findings and a positive urine culture. Patients were further classified according to the results of blood cultures into those with and without bacteremia. Plasma concentrations of sST2 were determined by a sensitive immunoassay. Non-parametric statistics were used for analysis.Results: The maternal plasma sST2 concentration increased with gestational age in normal pregnancies. Pregnant patients with acute pyelonephritis had a higher median (interquartile range) plasma sST2 concentration than those with a normal pregnancy [85 (47-239) ng/mL vs. 31 (14-52) ng/mL, p < .001]. Among patients with pyelonephritis, those with a positive blood culture had a median plasma concentration of sST2 higher than that of patients with a negative blood culture [258 (IQR: 75-305) ng/mL vs. 83 (IQR: 46-153) ng/mL; p = .03]. An elevated plasma concentration of sST2 ≥ 215 ng/mL had a sensitivity of 73% and a specificity of 95% (area under the receiver operating characteristic curve, 0.74; p = .003) with a positive likelihood ratio of 13.8 and a negative likelihood ratio of 0.3 for the identification of patients who had a positive blood culture.Conclusion: sST2 is a candidate biomarker to identify bacteremia in pregnant women with pyelonephritis. Rapid identification of these patients may optimize patient care.
Collapse
Affiliation(s)
- Carolyn Chatterton
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dahiana M. Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Gynecology and Obstetrics, Universidad del Valle, Cali, Colombia
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer Sheva, Israel
| | - Piya Chaemsaithong
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Kanninen T, Jung E, Gallo DM, Diaz-Primera R, Romero R, Gotsch F, Suksai M, Bosco M, Chaiworapongsa T. Soluble suppression of tumorigenicity-2 in pregnancy with a small-for-gestational-age fetus and with preeclampsia. J Matern Fetal Neonatal Med 2023; 36:2153034. [PMID: 36521862 PMCID: PMC10291739 DOI: 10.1080/14767058.2022.2153034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Preeclampsia and fetal growth disorders are pregnancy-specific conditions that share common pathophysiological mechanisms. Yet, why some patients develop preeclampsia while others experience fetal growth restriction, or a combination of both clinical presentations, is unknown. We propose that the difference in severity of the maternal inflammatory response can contribute to the clinical phenotypes of preeclampsia vs. small for gestational age (SGA). To assess this hypothesis, we measured maternal plasma concentrations of the soluble isoform of suppression of tumorigenicity-2 (sST2), a member of the interleukin-1 receptor family that buffers proinflammatory responses. Previous reports showed that serum sST2 concentrations rise in the presence of intravascular inflammation and Th1-type immune responses and are significantly higher in patients with preeclampsia compared to those with normal pregnancy. The behavior of sST2 in pregnancies complicated by SGA has not been reported. This study was conducted to compare sST2 plasma concentrations in normal pregnancies, in those with preeclampsia, and in those with an SGA fetus. METHODS This retrospective cross-sectional study included women with an SGA fetus (n = 52), women with preeclampsia (n = 106), and those with normal pregnancy (n = 131). Maternal plasma concentrations of sST2 were determined by enzyme-linked immunosorbent assay. Doppler velocimetry of the uterine and umbilical arteries was available in a subset of patients with SGA (42 patients and 43 patients, respectively). RESULTS (1) Women with an SGA fetus had a significantly higher median plasma concentration of sST2 than normal pregnant women (p = .008); (2) women with preeclampsia had a significantly higher median plasma concentration of sST2 than those with normal pregnancy (p < .001) and those with an SGA fetus (p < .001); (3) patients with SGA and abnormal uterine artery Doppler velocimetry had a higher median plasma concentration of sST2 than controls (p < .01) and those with SGA and normal uterine artery Doppler velocimetry (p = .02); (4) there was no significant difference in the median plasma sST2 concentration between patients with SGA who had normal uterine artery Doppler velocimetry and controls (p = .4); (5) among patients with SGA, those with abnormal and those with normal umbilical artery Doppler velocimetry had higher median plasma sST2 concentrations than controls (p = .001 and p = .02, respectively); and (6) there was no significant difference in the median plasma sST2 concentrations between patients with SGA who did and those who did not have abnormal umbilical artery Doppler velocimetry (p = .06). CONCLUSIONS Preeclampsia and disorders of fetal growth are conditions characterized by intravascular inflammation, as reflected by maternal plasma concentrations of sST2. The severity of intravascular inflammation is highest in patients with preeclampsia.
Collapse
Affiliation(s)
- Tomi Kanninen
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Gynecology and Obstetrics, Universidad del Valle, Cali, Colombia
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
11
|
Wang Y. Immune-related biomarkers in myocardial infarction; diagnostic/prognostic value and therapeutic potential. J Biochem Mol Toxicol 2023; 37:e23489. [PMID: 37574886 DOI: 10.1002/jbt.23489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/24/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
The incidence of myocardial infarction (MI) is increasing worldwide on an annual basis. The incorporation of circulating biomarkers, along with electrocardiography, echocardiography, coronary angiograms, and other diagnostic techniques, is essential in the evaluation, prediction, and therapeutic efficacy assessment of patients afflicted with MI. Biomarker evaluation has been employed in the diagnosis of MI for over five decades. Further biomarker research can be carried out as newer biomarkers have been discovered in pathways such as inflammatory response, neurohormonal stimulation, or myocardial stress that initiate significantly earlier than myocyte necrosis and the diagnostic establishment of cardiac troponins. The assessment of biomarkers for MI is on the brink of a significant transformation due to advancements in comprehending the intricate pathophysiology of the condition. This has led to a pursuit of innovative biomarkers that could potentially overcome the limitations of current biomarkers. For individuals with a high-risk profile, this may facilitate tailoring of appropriate treatment. This review places emphasis on a diverse array of biomarkers that have the potential to offer diagnostic and prognostic information, as well as the latest clinical and preclinical evidence that is driving theoretical advancements in cardiovascular immunotherapy.
Collapse
Affiliation(s)
- Yanhai Wang
- Clinical Laboratory Department, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
12
|
Witkowska A, Staciwa M, Duraj I, Wozniak E, Broncel M, Gorzelak-Pabis P. Interleukin-33/sST2: Dynamic assessment in patients with acute coronary syndrome. Adv Med Sci 2023; 68:195-201. [PMID: 37216709 DOI: 10.1016/j.advms.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE Interleukin (IL)-33 and its soluble receptor ST2 (sST2) play a crucial role in the immune response. sST2 has been approved by the Food and Drug Administration as a prognostic biomarker of mortality in chronic heart failure patients, however, the role of IL-33 and sST2 in atherosclerotic cardiovascular disease remains unclear. The aim of this study was to measure serum level of IL-33 and sST2 of patients at the onset of acute coronary syndrome (ACS) and 3 months after primary percutaneous revascularization. PATIENTS AND METHODS Forty patients were divided into ST segment elevation myocardial infarction (STEMI) group, non-ST segment elevation myocardial infarction (NSTEMI) and unstable angina (UA) group. IL-33 and sST2 level were measured with ELISA. Additionally, IL-33 expression in peripheral blood mononuclear cells (PBMCs), was evaluated. RESULTS All ACS patients had a significantly lower level of sST2 3 months after ACS as compared to the baseline (p < 0.039). The STEMI patients had higher serum levels of IL-33 at the moment of ACS as compared to 3 months after the event, with an average decrease of 17.87 pg/ml (p < 0.007). Conversely, sST2 serum levels were still high after 3 months following an ACS in STEMI patients. ROC curve demonstrated that increased IL-33 serum level could be STEMI predictor. CONCLUSIONS The assessment of the baseline and dynamics of changes in IL-33 and sST2 concentrations in patients with ACS may be important for the diagnostic process and may help in understanding of how the immune mechanisms work at the moment of an ACS event.
Collapse
Affiliation(s)
- Anna Witkowska
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland.
| | - Mateusz Staciwa
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Iwona Duraj
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Ewelina Wozniak
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Paulina Gorzelak-Pabis
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Alkhateeb A, Mahmoud HEM, AK M, Hassan MH, Muddathir ARM, Bakry AG. Impact of Myocardial Ischemia and Revascularization by Percutaneous Coronary Intervention on Circulating Level of Soluble ST2. Vasc Health Risk Manag 2023; 19:411-420. [PMID: 37434792 PMCID: PMC10332372 DOI: 10.2147/vhrm.s416206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Background The prognostic role of the soluble circulating suppression of tumorigenicity 2 marker (sST2) in different cardiovascular diseases (CVD) is still under investigation. This research aimed to assess the serum levels of sST2 in the blood of individuals with ischemic heart disease and its relation to disease severity, also to examine any changes in sST2 levels following a successful percutaneous coronary intervention (PCI) in those patients. Methods A total of 33 ischemic patients and 30 non-ischemic controls were included. The plasma level of sST2 was measured using commercially available ELISA assay kit, at baseline and 24-48 h after the intervention in the ischemic group. Results On admission, there was a significant difference between the group of acute/chronic coronary syndrome cases and controls regarding the sST2 plasma level (p < 0.001). There was an insignificant difference between the three ischemic subgroups at the baseline sST2 level (p = 0.38). The plasma sST2 level decreased significantly after PCI (from 20.70 ± 1.71 to 16.51 ± 2.43, p = 0.006). There was a modestly just significant positive correlation between the acute change in post-PCI sST2 level and the severity of ischemia as measured by the Modified Gensini Score (MGS) (r = 0.45, p = 0.05). In spite of the highly significant improvement in the coronary TIMI flow of ischemic group after PCI, there was insignificant negative correlation between the post- PCI delta change in the sST2 level and the post-PCI TIMI coronary flow grade. Conclusion A significantly high plasma level of sST2 in patients with myocardial ischemia and controlled cardiovascular risk factors showed an immediate reduction after successful revascularization. The high baseline level of the sST2 marker and the acute post-PCI reduction was mainly related to the severity of ischemia rather than left ventricular function.
Collapse
Affiliation(s)
- Areej Alkhateeb
- Cardiology Division of Internal Medicine Department, South Valley University Hospital, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| | - Hossam Eldin M Mahmoud
- Cardiology Division of Internal Medicine Department, South Valley University Hospital, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| | - Mohammed AK
- Cardiology Division of Internal Medicine Department, South Valley University Hospital, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| | - Mohammed H Hassan
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| | - Abdel Rahim Mahmoud Muddathir
- Department of Hematology and Blood Transfusion, Faculty of Medical Laboratory Science, Alzaeim Alazhari University, Khartoum, Sudan
| | - Ahmed G Bakry
- Cardiology Division of Internal Medicine Department, South Valley University Hospital, Faculty of Medicine, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
14
|
Brunetti G, Barile B, Nicchia GP, Onorati F, Luciani GB, Galeone A. The ST2/IL-33 Pathway in Adult and Paediatric Heart Disease and Transplantation. Biomedicines 2023; 11:1676. [PMID: 37371771 DOI: 10.3390/biomedicines11061676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
ST2 is a member of interleukin 1 receptor family with soluble sST2 and transmembrane ST2L isoforms. The ligand of ST2 is IL-33, which determines the activation of numerous intracytoplasmic mediators following the binding with ST2L and IL-1RAcP, leading to nuclear signal and cardiovascular effect. Differently, sST2 is released in the blood and works as a decoy receptor, binding IL-33 and blocking IL-33/ST2L interaction. sST2 is mainly involved in maintaining homeostasis and/or alterations of different tissues, as counterbalance/activation of IL-33/ST2L axis is typically involved in the development of fibrosis, tissue damage, inflammation and remodeling. sST2 has been described in different clinical reports as a fundamental prognostic marker in patients with cardiovascular disease, as well as marker for the treatment monitoring of patients with heart failure; however, further studies are needed to better elucidate its role. In this review we reported the current knowledge about its role in coronary artery disease, heart failure, heart transplantation, heart valve disease, pulmonary arterial hypertension, and cardiovascular interventions.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Francesco Onorati
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Antonella Galeone
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| |
Collapse
|
15
|
Søndergaard FT, Beske RP, Frydland M, Møller JE, Helgestad OKL, Jensen LO, Holmvang L, Goetze JP, Engstrøm T, Hassager C. Soluble ST2 in plasma is associated with post-procedural no-or-slow reflow after primary percutaneous coronary intervention in ST-elevation myocardial infarction. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2023; 12:48-52. [PMID: 36355574 DOI: 10.1093/ehjacc/zuac146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022]
Abstract
AIM The no-or-slow-reflow phenomenon after primary percutaneous coronary intervention is associated with more extensive myocardial injury in patients with ST-elevation myocardial infarction (STEMI). Soluble suppression of tumourigenicity 2 (sST2) is released in acute myocardial response to injury, and an increase in plasma level in the initial phase of STEMI is associated with increased mortality and risk of heart failure. We have therefore explored the association of pre-intervention plasma sST2 with the post-procedural no-or-slow-reflow phenomenon in patients with STEMI. METHODS AND RESULTS We included consecutive patients with verified STEMI from two tertiary heart centres. Blood samples were collected at admission before angiography. Post-procedural coronary flow was assessed according to thrombolysis in myocardial infarction (TIMI) classification for STEMI. Patients were divided into two groups: post-procedural TIMI 0-2 as no-or-slow reflow and TIMI 3 as normal reflow. The association between sST2 and TIMI flow was explored using multiple logistic regression. A total of 1607 patients with available TIMI flow classification were included in the analysis. Normal reflow was seen in 1520 (94.6%), while 87 (5.4%) had no-or-slow reflow. No-or-slow-reflow patients had higher all-cause 30-day mortality [10 (11%) vs. 65 (4.3%), P = 0.006]. Pre-procedural sST2 was higher in the no-or-slow-flow group [47 ng/mL, interquartile range (IQR, 33-83) vs. 39 ng/mL (IQR 29-55), P < 0.001] and was independently associated with post-procedural no-or-slow flow [two-fold sST2 increase: odds ratio 1.44 (1.15-1.78), P = 0.0012]. CONCLUSION In patients with STEMI, the sST2 level at admission before coronary angiography is independently associated with the post-procedural no-or-slow-reflow phenomenon.
Collapse
Affiliation(s)
- Frederik T Søndergaard
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Rasmus P Beske
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Martin Frydland
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Jacob Eifer Møller
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark.,Department of Cardiology-B, Odense University Hospital, Odense, Denmark
| | - Ole K L Helgestad
- Department of Cardiology-B, Odense University Hospital, Odense, Denmark
| | - Lisette Okkels Jensen
- Department of Cardiology-B, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Odense University Hospital, Odense, Denmark
| | - Lene Holmvang
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| | - Christian Hassager
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej, Copenhagen 9 DK-2100, Denmark
| |
Collapse
|
16
|
Aispuru-Lanche GR, Gallego-Muñoz M, Jayo-Montoya JA, Villar-Zabala B, Maldonado-Martín S. Low-Volume and High-Intensity Aerobic Interval Training May Attenuate Dysfunctional Ventricular Remodeling after Myocardial Infarction: Data from the INTERFARCT Study. Rev Cardiovasc Med 2023; 24:20. [PMID: 39076876 PMCID: PMC11270403 DOI: 10.31083/j.rcm2401020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 07/31/2024] Open
Abstract
Background Aerobic high-intensity interval training (HIIT) has demonstrated benefits for ventricular remodeling after myocardial infarction (MI) through various mechanisms. Despite this, the optimal training volume is not well known. The present study aimed to assess the effects of different (low vs. high volume) aerobic HIIT compared to an attentional control (AC) group on echocardiographic and biochemical indicators of left ventricular (LV) remodeling in adults after MI. Methods Randomized clinical trial conducted on post-MI patients with preserved ventricular function. Participants were assigned to three study groups. Two groups performed HIIT 2 d/week, one group with low-volume HIIT (20 min, n = 28) and another with high-volume HIIT (40 min, n = 28). A third group was assigned to AC (n = 24) with recommendations for unsupervised aerobic training. Left ventricular echocardiographic parameters and cardiac biomarker levels (N-terminal pro-b-type natriuretic peptide, NT-proBNP; soluble growth stimulation expressed gene 2, ST2; troponin T; and creatine kinase) were assessed at baseline and after the intervention (16 weeks). Results Eighty participants (58.4 ± 8.3 yrs, 82.5% male) were included. Both low- and high-volume HIIT showed increases (p < 0.05) in left ventricular end-diastolic diameter (1.2%, 2.6%), and volume (1.1%, 1.3%), respectively. Interventricular septal and posterior walls maintained their thickness (p = 0.36) concerning the AC. Significant (p < 0.05) gain in diastolic function was shown with the improvements in E (-2.1%, -3.3%), e' waves (2.2%, 5.5%), and the deceleration time (2.1%, 2.9%), and in systolic function with a reduction in global longitudinal strain (-3.2%, -4.7%), respectively. Significant (p < 0.05) reductions of N-terminal pro-B-type natriuretic peptide (NT-proBNP) (-4.8%, -11.1%) and of ST2 (-21.7%, -16.7%)were found in both HIIT groups respectively compared to the AC group. Creatine kinase elevation was shown only in high-volume HIIT (19.3%, p < 0.01). Conclusions Low-volume HIIT is proposed as a clinically time-efficient and safer strategy to attenuate dysfunctional remodeling by preventing wall thinning and improving LV function in post-MI patients.
Collapse
Affiliation(s)
- Gualberto Rodrigo Aispuru-Lanche
- Department of Physiology, Faculty of Medicine, University of the Basque Country (UPV/EHU), Leioa, 48940 Bizkaia, Basque Country, Spain
- Primary Care Administration of Burgos, Salud Castilla y Leon (Sacyl), 09267 Burgos, Spain
| | - Monica Gallego-Muñoz
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, 01006 Alava/Araba, Basque Country, Spain
| | - Jon Ander Jayo-Montoya
- Department of Physical Education and Sport, Faculty of Education and Sport, Physical Activity and Sport Sciences Section, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Alava, Basque Country, Spain
| | - Beatriz Villar-Zabala
- Primary Care Administration of Burgos, Salud Castilla y Leon (Sacyl), 09267 Burgos, Spain
| | - Sara Maldonado-Martín
- GIzartea, Kirola eta Ariketa Fisikoa Ikerkuntza Taldea (GIKAFIT), Society, Sports, and Physical Exercise Research Group, Department of Physical Education and Sport, Faculty of Education and Sport-Physical Activity and Sport Sciences Section, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, 01007 Araba/Álava, Basque Country, Spain
- Bioaraba Health Research Institute, Physical Activity, Exercise, and Health group, 01007 Vitoria-Gasteiz, Basque Country, Spain
| |
Collapse
|
17
|
Kreiniz N, Gertz MA. Bad players in AL amyloidosis in the current era of treatment. Expert Rev Hematol 2023; 16:33-49. [PMID: 36620914 PMCID: PMC9905376 DOI: 10.1080/17474086.2023.2166924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Systemic AL amyloidosis (ALA) is a clonal plasma cell (PC) disease characterized by deposition of amyloid fibrils in different organs and tissues. Traditionally, the prognosis of ALA is poor and is primarily defined by cardiac involvement. The modern prognostic models are based on cardiac markers and free light chain difference (dFLC). Cardiac biomarkers have low specificity and are dependent on renal function, volume status, and cardiac diseases other than ALA. New therapies significantly improved the prognosis of the disease. The advancements in technologies - cardiac echocardiography (ECHO) and cardiac MRI (CMR), as well as new biological markers, relying on cardiac injury, inflammation, endothelial damage, and clonal and non-clonal PC markers are promising. AREAS COVERED An update on the prognostic significance of cardiac ALA, number of involved organs, response to treatment, including minimal residual disease (MRD), ECHO, MRI, and new biological markers will be discussed. The literature search was done in PubMed and Google Scholar, and the most recent and relevant data are included. EXPERT OPINION Prospective multicenter trials, evaluating multiple clinical and laboratory parameters, should be done to improve the risk assessment models in ALA in the modern era of therapy.
Collapse
Affiliation(s)
- Natalia Kreiniz
- Division of Hematology, Bnai Zion Medical Centre, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | |
Collapse
|
18
|
Prediction of one-year adverse clinical outcomes by macrophage migration inhibitory factor in stemi patients. EUREKA: HEALTH SCIENCES 2022. [DOI: 10.21303/2504-5679.2022.002714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Biomarkers have taken one of the first places as diagnostic and prognostic tools in ST-segment elevation myocardial infarction (STEMI) and are consequently widely used as predictors of short-term and long-term prognosis. One of the promising biomarkers for early cardiovascular outcomes prediction is the pro-inflammatory cytokine macrophage migration inhibitory factor (MIF).
The aim of the study was to elucidate a plausible predictive value of the MIF levels for one-year clinical outcomes in STEMI patients who underwent primary percutaneous coronary intervention (PCI).
Materials and methods. 134 STEMI patients were enrolled in the study after receiving voluntary informed consent. All patients underwent conventional investigations, and additionally, the MIF levels were determined at baseline, directly before and after PCI. During 1-year follow-up, 37 % of patients reached the endpoint, which was composite and included all-cause mortality, non-fatal myocardial infarction, non-fatal stroke, hospitalization for unstable angina, heart failure decompensation, and urgent revascularization.
Results. We have found that pre-PCI MIF levels > 3934 pg/mL (AUC=0.7; 95 % CI 0.578 to 0.753; Youden index=0.31; p=0.008) might be an independent predictor of composite endpoints with sensitivity 54 % and specificity 82 %. A positive correlation between MIF and inflammatory biomarkers was revealed (WBC count r=0.33, p=0.0001; CRP r=0.19, p=0.032). Adverse outcomes associated with higher pre- and post-PCI MIF levels (OR 1.0, 95 % CI 1.0001–1.0008; p=0.013 and OR 1.0, 95 % CI 1.0001–1.0009; p=0.019) and CRP that determined during the first week after the event (OR 1.0, 95 % CI 1.005–1.2, p=0.03). Kaplan-Meier analysis has shown a substantially lower long-term survival rate in patients with a MIF level > 3493 pg/ml compared to a MIF level ≤ 3493 pg/ml (Log rank=0.00025).
Conclusions. The MIF levels exceeding 3934 ng/ml were associated with a higher risk of one-year adverse clinical outcomes in STEMI patients who underwent primary PCI.
Collapse
|
19
|
Zhang J, Chen Z, Ma M, He Y. Soluble ST2 in coronary artery disease: Clinical biomarkers and treatment guidance. Front Cardiovasc Med 2022; 9:924461. [PMID: 36225958 PMCID: PMC9548599 DOI: 10.3389/fcvm.2022.924461] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The IL-33/ST2 L signaling pathway is involved in the pathophysiological processes of several diseases and mainly exerts anti-inflammatory and antifibrotic effects. Soluble suppression of tumorigenicity 2 (sST2), which serves as a competitive inhibitory molecule of this pathway, is a member of the interleukin (IL)-1 family, a decoy receptor for IL33, thought to play a role in cardiac remodeling and the inflammatory process. However, the association between sST2 and coronary artery disease (CAD), one of the most common causes of heart failure, is still being explored. We therefore reviewed the research on sST2 in the field of CAD, including reflecting the atherosclerosis burden, predicting no-reflow, predicting prognosis, responding to myocardial remodeling, and guiding management, hoping to provide cardiologists with new perspectives.
Collapse
|
20
|
The Octopus Trap of Takotsubo and Stroke: Genetics, Biomarkers and Clinical Management. J Pers Med 2022; 12:jpm12081244. [PMID: 36013193 PMCID: PMC9410002 DOI: 10.3390/jpm12081244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Takotsubo cardiomyopathy (TC) is a reversible cardiomyopathy mimicking an acute coronary syndrome, usually observed in response to acute stress situations. The association between acute ischemic stroke and TC is already known, since it has been previously reported that ischemic stroke can be both a consequence and a potential cause of TC. However, the precise pathophysiological mechanism linking the two conditions is still poorly understood. The aim of our review is to expand insights regarding the genetic susceptibility and available specific biomarkers of TC and to investigate the clinical profile and outcomes of patients with TC and stroke. Since evidence and trials on TC and stroke are currently lacking, this paper aims to fill a substantial gap in the literature about the relationship between these pathologies.
Collapse
|
21
|
Leancă SA, Crișu D, Petriș AO, Afrăsânie I, Genes A, Costache AD, Tesloianu DN, Costache II. Left Ventricular Remodeling after Myocardial Infarction: From Physiopathology to Treatment. Life (Basel) 2022; 12:1111. [PMID: 35892913 PMCID: PMC9332014 DOI: 10.3390/life12081111] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of death and morbidity worldwide, with an incidence relatively high in developed countries and rapidly growing in developing countries. The most common cause of MI is the rupture of an atherosclerotic plaque with subsequent thrombotic occlusion in the coronary circulation. This causes cardiomyocyte death and myocardial necrosis, with subsequent inflammation and fibrosis. Current therapies aim to restore coronary flow by thrombus dissolution with pharmaceutical treatment and/or intravascular stent implantation and to counteract neurohormonal activation. Despite these therapies, the injury caused by myocardial ischemia leads to left ventricular remodeling; this process involves changes in cardiac geometry, dimension and function and eventually progression to heart failure (HF). This review describes the pathophysiological mechanism that leads to cardiac remodeling and the therapeutic strategies with a role in slowing the progression of remodeling and improving cardiac structure and function.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Daniela Crișu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antoniu Octavian Petriș
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| | - Irina Afrăsânie
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Antonia Genes
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dan Nicolae Tesloianu
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
| | - Irina Iuliana Costache
- Department of Cardiology, Emergency Clinical Hospital “Sf. Spiridon”, Bd. Independentei nr. 1, 700111 Iasi, Romania; (S.A.L.); (A.O.P.); (I.A.); (A.G.); (D.N.T.); (I.I.C.)
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Str. University nr. 16, 700083 Iasi, Romania;
| |
Collapse
|
22
|
Melo APC, Teixeira HMP, Coelho RS, De Jesus TDS, Queiroz GA, Silva HDS, De Almeida YCF, Alcantara-Neves NM, De Matos SMA, D'innocenzo S, Silva RDCR, Lima Barreto M, Costa RDS, Pinto LC, Figueiredo CA. Variants in proinflammatory genes IL1RL1, IL1B and IRF4 are associated with overweight in a pediatric Brazilian population. Gene X 2022; 828:146478. [PMID: 35390444 DOI: 10.1016/j.gene.2022.146478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Obesity is a chronic complex disease with great prevalence for children all over the world. Characterized for low-grade inflammation associated with several comorbidities such as resistance and type 2 diabetes mellitus (T2DM). OBJECTIVES To investigate whether genetic variants in IL10, IL1RL1, IL1B, IRF4, TNF, IL6, and IL33 genes are associated with being overweight in children. METHODS We performed the genotyping of 1004 children using Illumina 2.5 Human Omni bead chip, and association analysis on the genetic variants and the overweight through logistic regression adjusted for sex, age and components principal. RESULTS Of the seven genes analyzed, 16 SNVs significantly associated. Eleven variants in IL1RL1, two in IL1B and one in IRF4 genes increased overweight risk and two SNVs in IL1RL1 were associated with protection against overweight. The rs2287047-A was negatively associated (OR: 0.66, CI95%: 0.19-0.45) and had a reduced IL1RL1 expression in whole blood (p 0.033) in silico eQTL. The rs12203592-T, in IRF4, was positively associated with being overweight, and led to an increased gene expression in whole blood (p < 0.001) and adipose tissue (p < 0.001). CONCLUSION These results suggest that genetic variants in inflammatory genes may play an important role in the development of overweight in children.
Collapse
Affiliation(s)
| | | | - Raisa Santos Coelho
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, BA, Brazil
| | | | | | | | | | | | | | - Silvana D'innocenzo
- Instituto de Saúde Coletiva, Universidade Federal Da Bahia, Salvador, BA, Brazil
| | | | - Maurício Lima Barreto
- Instituto de Saúde Coletiva, Universidade Federal Da Bahia, Salvador, BA, Brazil; CIDACS - Centro de Integração De Dados E Conhecimentos Para Saúde, Fiocruz, Brazil
| | | | | | | |
Collapse
|
23
|
Hammer F, Genser B, Dieplinger B, Egger M, Müller T, Drechsler C, März W, Störk S, Wanner C, Krane V. Soluble suppression of tumorigenesis-2 (sST2) is a strong predictor of all-cause, cardiovascular and infection-related mortality risk in hemodialysis patients with diabetes mellitus. Clin Kidney J 2022; 15:1915-1923. [PMID: 36158148 PMCID: PMC9494540 DOI: 10.1093/ckj/sfac142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background Soluble suppression of tumorigenesis-2 (sST2) is a strong prognostic biomarker of cardiovascular (CV) disease. End-stage kidney disease (ESKD) patients are at high risk of CV events and infections. Herein we investigated the utility of sST2 to predict all-cause and cause-specific mortality in haemodialysis (HD) patients with diabetes mellitus. Methods sST2 concentrations were measured in plasma samples of 1196 participants of the German Diabetes and Dialysis (4D) study who had type 2 diabetes mellitus and received maintenance HD for ESKD. Hazard ratios (HRs) for prespecified, adjudicated endpoints were determined according to sST2 levels at baseline by multivariate Cox proportional hazards analysis. Results Participants (mean age 66 years, 54% male) had a median sST2 concentration of 25 ng/mL and were followed up for 4 years. After adjustment for possible confounders, participants with sST2 concentrations in the highest (>32.6 ng/mL) compared with the lowest (<20.1 ng/mL) quartile exhibited a 2-fold higher all-cause mortality risk {[HR 2.06 95% confidence interval (CI) 1.61–2.61]; P < .001}. High sST concentrations (fourth versus first quartile) were strongly associated with the risk of cardiac death [HR 2.29 (95% CI 1.55–3.39); P < .001]. Analysis of individual components of cardiac causes of death showed an increased risk of sudden death [HR 2.24 (95% CI 1.33–3.77); P < .001], death due to myocardial infarction [HR 2.12 (95% CI 0.9–5.0); P = .087] and heart failure [HR 3.34 (95% CI 1.15–9.75); P = .027] in participants with sST2 levels in the highest compared with the lowest quartile. Likewise, participants with the highest sST2 levels had an increased risk of fatal stroke [HR 1.92 (95% CI 1.17–3.14); P = .009] and fatal infections [HR 2.01 (95% CI 1.2–3.37); P = .008]. In contrast to fatal CV events, sST2 was not associated with the risk of non-fatal myocardial infarction [HR 0.68 (95% CI 0.41–1.12); P = .132] or non-fatal stroke [HR 1.28 (95% CI 0.64–2.53); P = .485]. Conclusions In HD patients with diabetes mellitus, high concentrations of sST2 were strongly and independently associated with an increased risk of all-cause mortality, CV mortality and death due to infection but not non-fatal CV events.
Collapse
Affiliation(s)
- Fabian Hammer
- Department of Internal Medicine B, Division of Cardiology, University Medicine Greifswald, Greifswald, Germany
- Department of Internal Medicine I, Division of Cardiology, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Bernd Genser
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), Medical Faculty Mannheim, Heidelberg University
- BGStats Consulting, Vienna, Austria
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Margot Egger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Thomas Müller
- Department of Laboratory Diagnostics, Hospital of Gmunden, Austria
| | - Christiane Drechsler
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - Winfried März
- Synlab Akademie für ärztliche Fortbildung, Synlab Services GmbH, Mannheim, Germany
| | - Stefan Störk
- Department of Internal Medicine I, Division of Cardiology, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Wanner
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - Vera Krane
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Urban MH, Stojkovic S, Demyanets S, Hengstenberg C, Valipour A, Wojta J, Burghuber OC. Soluble ST2 and All-Cause Mortality in Patients with Chronic Obstructive Pulmonary Disease—A 10-Year Cohort Study. J Clin Med 2021; 11:jcm11010056. [PMID: 35011794 PMCID: PMC8745630 DOI: 10.3390/jcm11010056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory condition with constantly increasing mortality rates. Interleukin (IL)-33 and its decoy receptor, soluble suppression of tumorigenicity 2 (sST2), play a central role in the inflammatory response during infection. sST2 was suggested as a factor in the pathogenesis of COPD and emerged as a predictor of mortality in other non-communicable diseases. The role of sST2 as a predictor of mortality remains unclear in COPD yet. In this cohort study, we measured circulating concentrations of IL-33 and sST2 in the serum of patients with stable COPD (n = 59), patients with acute exacerbation of COPD (n = 29) and smoking (n = 20) and non-smoking controls (n = 20), using commercially available ELISAs, and investigated the prognostic role of sST2 in stable COPD. sST2 levels were significantly higher in COPD patients and smokers compared with non-smoking controls. We identified systolic blood pressure, forced expiratory volume in 1 s (FEV1% predicted), neutrophil count, lactate dehydrogenase and pack-years index as independent predictors of sST2 levels. During a median follow-up time of 10.6 years, 28 patients (47.5%) died. sST2 was an independent predictor of all-cause mortality in patients with COPD with a hazard ratio of 2.9 (95% CI 1.1–8.4, p = 0.035) per one standard deviation after adjustment for age, sex, pack-years, FEV1% predicted and C-reactive protein (CRP). sST2 concentrations are associated with severity of disease and long-term outcome in patients with COPD.
Collapse
Affiliation(s)
- Matthias H. Urban
- Department of Internal and Respiratory Medicine and Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Klinik Floridsdorf, 1210 Vienna, Austria; (M.H.U.); (A.V.)
- Ludwig Boltzmann Institute for Lung Health, 1140 Vienna, Austria;
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (S.S.); (C.H.)
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christian Hengstenberg
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (S.S.); (C.H.)
| | - Arschang Valipour
- Department of Internal and Respiratory Medicine and Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Klinik Floridsdorf, 1210 Vienna, Austria; (M.H.U.); (A.V.)
| | - Johann Wojta
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (S.S.); (C.H.)
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
- Core Facilities, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +431-404-007-3500
| | - Otto C. Burghuber
- Ludwig Boltzmann Institute for Lung Health, 1140 Vienna, Austria;
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
| |
Collapse
|
25
|
Fotiou D, Theodorakakou F, Kastritis E. Biomarkers in AL Amyloidosis. Int J Mol Sci 2021; 22:ijms222010916. [PMID: 34681575 PMCID: PMC8536050 DOI: 10.3390/ijms222010916] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/02/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023] Open
Abstract
Systemic AL amyloidosis is a rare complex hematological disorder caused by clonal plasma cells which produce amyloidogenic immunoglobulins. Outcome and prognosis is the combinatory result of the extent and pattern of organ involvement secondary to amyloid fibril deposition and the biology and burden of the underlying plasma cell clone. Prognosis, as assessed by overall survival, and early outcomes is determined by degree of cardiac dysfunction and current staging systems are based on biomarkers that reflect the degree of cardiac damage. The risk of progression to end-stage renal disease requiring dialysis is assessed by renal staging systems. Longer-term survival and response to treatment is affected by markers of the underlying plasma cell clone; the genetic background of the clonal disease as evaluated by interphase fluorescence in situ hybridization in particular has predictive value and may guide treatment selection. Free light chain assessment forms the basis of hematological response criteria and minimal residual disease as assessed by sensitive methods is gradually being incorporated into clinical practice. However, sensitive biomarkers that could aid in the early diagnosis and that could reflect all aspects of organ damage and disease biology are needed and efforts to identify them are continuous.
Collapse
|
26
|
Association of soluble ST2 and infarct location within 12-24 h in STEMI: A cross-sectional study. Ann Med Surg (Lond) 2021; 70:102844. [PMID: 34540221 PMCID: PMC8435924 DOI: 10.1016/j.amsu.2021.102844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Background ST-Segment Elevation Myocardial Infarction (STEMI) causes the release of soluble ST2 biomarkers at high level on acute phase. However, sST2 has never been used as adjunct biomarker in ESC/AHA guideline for STEMI. Furthermore, the specific onset that sST2 may have role in acute phase of STEMI related with infarct location has not been established. This study aimed to prove the association between serum ST2 levels and infarct location in STEMI. Material and methods This study was cross-sectional. STEMI patients with onset of anginal pain 12–24 h were included in study. The exclusion criterias were patients with AMI aside from STEMI and other potential confounders affecting the sST2 level. Serum sST2 was collected on first medical contact when admitted to emergency unit. The patients were grouped into anterior STEMI and non-anterior STEMI. sST2 levels were compared with demographics data, clinical and laboratory variables using Student's t-test. Correlation of sST2 levels was analyzed using Spearman's correlation coefficient. Results 19 subjects were included in the anterior STEMI and 20 subjects were included in the non-anterior STEMI. We found no difference in sST2 levels between anterior STEMI and non-anterior STEMI (mean ± SD; 729.97 pg/mL ± 147.78 pg/mL vs 606.87 pg/mL ± 147.78 pg/mL, p = 0.119). Onset was correlated with serum sST2 levels in male subjects (r = −0.459, p = 0.012). We found significant difference of mean sST2 between 2 onset groups divided at median (12–18 h vs 19–24 h, Δ mean = 107.75 pg/mL, p-value = 0.021). Conclusion sST2 was not associated with infarct location within 12–24 h onset of STEMI. This results suggest that infarct location might not responsible for the elevation of serum sST2 levels in acute phase of STEMI. STEMI causes the release of soluble ST2 biomarkers at high level on acute phase. sST2 and it association with infarct location within 12–24 h. Infarct location might not responsible for the elevation of serum sST2 levels.
Collapse
|
27
|
Ambari AM, Setianto B, Santoso A, Radi B, Dwiputra B, Susilowati E, Tulrahmi F, Wind A, Cramer MJM, Doevendans P. Randomised controlled trial into the role of ramipril in fibrosis reduction in rheumatic heart disease: the RamiRHeD trial protocol. BMJ Open 2021; 11:e048016. [PMID: 34518254 PMCID: PMC8438922 DOI: 10.1136/bmjopen-2020-048016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rheumatic heart disease (RHD) is a major burden in developing countries and accounts for 80% of all people living with the disease, where it causes most cardiovascular morbidity and mortality in children and young adults. Chronic inflammation and fibrosis of heart valve tissue due to chronic inflammation in RHD will cause calcification and thickening of the impacted heart valves, especially the mitral valve. This fibrogenesis is enhanced by the production of angiotensin II by increased transforming growth factor β expression and later by the binding of interleukin-33, which is known to have antihypertrophic and antifibrotic effects, to soluble sST2. sST2 binding to this non-natural ligand worsens fibrosis. Therefore, we hypothesise that ACE inhibitors (ACEIs) would improve rheumatic mitral valve stenosis. METHODS AND ANALYSIS This is a single-centre, double-blind, placebo-controlled, randomised clinical trial with a pre-post test design. Patients with rheumatic mitral stenosis and valve dysfunction will be planned for cardiac valve replacement operation and will be given ramipril 5 mg or placebo for a minimum of 12 weeks before the surgery. The expression of ST2 in the mitral valve is considered to be representative of cardiac fibrosis. Mitral valve tissue will be stained by immunohistochemistry to ST2. Plasma ST2 will be measured by ELISA. This study is conducted in the Department of Cardiology and Vascular Medicine, Universitas Indonesia, National Cardiac Center Harapan Kita Hospital, Jakarta, Indonesia, starting on 27 June 2019. ETHICS AND DISSEMINATION The performance and dissemination of this study were approved by the ethics committee of National Cardiovascular Center Harapan Kita with ethical code LB.02.01/VII/286/KEP.009/2018. TRIAL REGISTRATION NUMBER NCT03991910.
Collapse
Affiliation(s)
- Ade Meidian Ambari
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Budhi Setianto
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Anwar Santoso
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Basuni Radi
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Bambang Dwiputra
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Eliana Susilowati
- Research Assistant of Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
| | - Fadilla Tulrahmi
- Research Assistant of Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
| | - Annemiek Wind
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Pieter Doevendans
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Central Military Hospital, Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
28
|
Merino-Merino A, Gonzalez-Bernal J, Fernandez-Zoppino D, Saez-Maleta R, Perez-Rivera JA. The Role of Galectin-3 and ST2 in Cardiology: A Short Review. Biomolecules 2021; 11:1167. [PMID: 34439833 PMCID: PMC8393977 DOI: 10.3390/biom11081167] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 01/21/2023] Open
Abstract
Galectin-3 is a lectin that binds beta-galactosides. It is involved in cardiac remodeling and fibrosis through the activation of macrophages and fibroblasts. ST2 is secreted by myocardial cells due to cardiac overload. These two biomarkers have been traditionally studied in the field of heart failure to guide medical therapy and detect the progression of the disease. Nevertheless, there are novel evidences that connect galectin-3 and ST2 with coronary heart disease and, specifically, with atrial fibrillation. The aim of this article is to concisely review the diagnostic and prognostic role of galectin-3 and ST2 in different cardiac diseases.
Collapse
Affiliation(s)
- Ana Merino-Merino
- Department of Cardiology, University Hospital of Burgos, 09005 Burgos, Spain;
| | | | - Dario Fernandez-Zoppino
- Department of Health Sciences, University of Burgos, 09005 Burgos, Spain; (J.G.-B.); (D.F.-Z.)
- National Scientific and Technical Research Council (CONICET), 2290 Buenos Aires, Argentina
| | - Ruth Saez-Maleta
- Department Clinical Analysis, University Hospital of Burgos, 09005 Burgos, Spain;
| | | |
Collapse
|
29
|
Alladina J, Levy SD, Cho JL, Brait KL, Rao SR, Camacho A, Hibbert KA, Harris RS, Medoff BD, Januzzi JL, Thompson BT, Bajwa EK. Plasma Soluble Suppression of Tumorigenicity-2 Associates with Ventilator Liberation in Acute Hypoxemic Respiratory Failure. Am J Respir Crit Care Med 2021; 203:1257-1265. [PMID: 33400890 DOI: 10.1164/rccm.202005-1951oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Rationale: Standard physiologic assessments of extubation readiness in patients with acute hypoxemic respiratory failure (AHRF) may not reflect lung injury resolution and could adversely affect clinical decision-making and patient outcomes. Objectives: We hypothesized that elevations in inflammatory plasma biomarkers sST2 (soluble suppression of tumorigenicity-2) and IL-6 indicate ongoing lung injury in AHRF and better inform patient outcomes compared with standard clinical assessments. Methods: We measured daily plasma biomarkers and physiologic variables in 200 patients with AHRF for up to 9 days after intubation. We tested the associations of baseline values with the primary outcome of unassisted breathing at Day 29. We analyzed the ability of serial biomarker measurements to inform successful ventilator liberation. Measurements and Main Results: Baseline sST2 concentrations were higher in patients dead or mechanically ventilated versus breathing unassisted at Day 29 (491.7 ng/ml [interquartile range (IQR), 294.5-670.1 ng/ml] vs. 314.4 ng/ml [IQR, 127.5-550.1 ng/ml]; P = 0.0003). Higher sST2 concentrations over time were associated with a decreased probability of ventilator liberation (hazard ratio, 0.80 per log-unit increase; 95% confidence interval [CI], 0.75-0.83; P = 0.03). Patients with higher sST2 concentrations on the day of liberation were more likely to fail liberation compared with patients who remained successfully liberated (320.9 ng/ml [IQR, 181.1- 495.6 ng/ml] vs. 161.6 ng/ml [IQR, 95.8-292.5 ng/ml]; P = 0.002). Elevated sST2 concentrations on the day of liberation decreased the odds of successful liberation when adjusted for standard physiologic parameters (odds ratio, 0.325; 95% CI, 0.119-0.885; P = 0.03). IL-6 concentrations did not associate with outcomes. Conclusions: Using sST2 concentrations to guide ventilator management may more accurately reflect underlying lung injury and outperform traditional measures of readiness for ventilator liberation.
Collapse
Affiliation(s)
| | - Sean D Levy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Josalyn L Cho
- Division of Pulmonary, Critical Care, and Occupational Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | | - Sowmya R Rao
- Boston University School of Public Health, Boston, Massachusetts; and
| | - Alexander Camacho
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
| | | | - R Scott Harris
- Division of Pulmonary and Critical Care Medicine and.,Vertex Pharmaceuticals, Boston, Massachusetts
| | | | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Ednan K Bajwa
- Division of Pulmonary and Critical Care Medicine and
| |
Collapse
|
30
|
van den Berg VJ, Umans VAWM, Brankovic M, Oemrawsingh RM, Asselbergs FW, van der Harst P, Hoefer IE, Kietselaer B, Crijns HJGM, Lenderink T, Oude Ophuis AJ, van Schaik RH, Kardys I, Boersma E, Akkerhuis KM. Stabilization patterns and variability of hs-CRP, NT-proBNP and ST2 during 1 year after acute coronary syndrome admission: results of the BIOMArCS study. Clin Chem Lab Med 2021; 58:2099-2106. [PMID: 32383686 DOI: 10.1515/cclm-2019-1320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/13/2020] [Indexed: 11/15/2022]
Abstract
Objectives Details of the biological variability of high-sensitivity C-reactive protein (hs-CRP), N-terminal prohormone of brain natriuretic peptide (NT-proBNP) and ST2 are currently lacking in patients with acute coronary syndrome (ACS) but are crucial knowledge when aiming to use these biomarkers for personalized risk prediction. In the current study, we report post-ACS kinetics and the variability of the hs-CRP, NT-proBNP and ST2. Methods BIOMArCS is a prospective, observational study with high frequency blood sampling during 1 year post-ACS. Using 1507 blood samples from 191 patients that remained free from adverse cardiac events, we investigated post-ACS kinetics of hs-CRP, NT-proBNP and ST2. Biological variability was studied using the samples collected between 6 and 12 months after the index ACS, when patients were considered to have stable coronary artery disease. Results On average, hs-CRP rose peaked at day 2 and rose well above the reference value. ST2 peaked immediately after the ACS but never rose above the reference value. NT-proBNP level rose on average during the first 2 days post-ACS and slowly declined afterwards. The within-subject variation and relative change value (RCV) of ST2 were relatively small (13.8%, RCV 39.7%), while hs-CRP (41.9%, lognormal RCV 206.1/-67.3%) and NT-proBNP (39.0%, lognormal RCV 185.2/-64.9%) showed a considerable variation. Conclusions Variability of hs-CRP and NT-proBNP within asymptomatic and clinically stable post-ACS patients is considerable. In contrast, within-patient variability of ST2 is low. Given the low within-subject variation, ST2 might be the most useful biomarker for personalizing risk prediction in stable post-ACS patients.
Collapse
Affiliation(s)
- Victor J van den Berg
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands.,Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | | | - Milos Brankovic
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands
| | - Rohit M Oemrawsingh
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands
| | - Folkert W Asselbergs
- Netherlands Heart Institute, Utrecht, The Netherlands.,University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pim van der Harst
- Netherlands Heart Institute, Utrecht, The Netherlands.,University Medical Center Groningen, Groningen, The Netherlands
| | - Imo E Hoefer
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | - Anton J Oude Ophuis
- Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands.,Working Group on Cardiovascular Research the Netherlands (WCN), Utrecht, The Netherlands
| | - Ron H van Schaik
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands
| | - Isabella Kardys
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands
| | - Eric Boersma
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands.,Erasmus MC, Department of Cardiology, Room Na 342, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Erasmus University Medical Center and Cardiovascular Research Institute COEUR, Rotterdam, The Netherlands
| | | |
Collapse
|
31
|
Sikorski V, Karjalainen P, Blokhina D, Oksaharju K, Khan J, Katayama S, Rajala H, Suihko S, Tuohinen S, Teittinen K, Nummi A, Nykänen A, Eskin A, Stark C, Biancari F, Kiss J, Simpanen J, Ropponen J, Lemström K, Savinainen K, Lalowski M, Kaarne M, Jormalainen M, Elomaa O, Koivisto P, Raivio P, Bäckström P, Dahlbacka S, Syrjälä S, Vainikka T, Vähäsilta T, Tuncbag N, Karelson M, Mervaala E, Juvonen T, Laine M, Laurikka J, Vento A, Kankuri E. Epitranscriptomics of Ischemic Heart Disease-The IHD-EPITRAN Study Design and Objectives. Int J Mol Sci 2021; 22:6630. [PMID: 34205699 PMCID: PMC8235045 DOI: 10.3390/ijms22126630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Epitranscriptomic modifications in RNA can dramatically alter the way our genetic code is deciphered. Cells utilize these modifications not only to maintain physiological processes, but also to respond to extracellular cues and various stressors. Most often, adenosine residues in RNA are targeted, and result in modifications including methylation and deamination. Such modified residues as N-6-methyl-adenosine (m6A) and inosine, respectively, have been associated with cardiovascular diseases, and contribute to disease pathologies. The Ischemic Heart Disease Epitranscriptomics and Biomarkers (IHD-EPITRAN) study aims to provide a more comprehensive understanding to their nature and role in cardiovascular pathology. The study hypothesis is that pathological features of IHD are mirrored in the blood epitranscriptome. The IHD-EPITRAN study focuses on m6A and A-to-I modifications of RNA. Patients are recruited from four cohorts: (I) patients with IHD and myocardial infarction undergoing urgent revascularization; (II) patients with stable IHD undergoing coronary artery bypass grafting; (III) controls without coronary obstructions undergoing valve replacement due to aortic stenosis and (IV) controls with healthy coronaries verified by computed tomography. The abundance and distribution of m6A and A-to-I modifications in blood RNA are charted by quantitative and qualitative methods. Selected other modified nucleosides as well as IHD candidate protein and metabolic biomarkers are measured for reference. The results of the IHD-EPITRAN study can be expected to enable identification of epitranscriptomic IHD biomarker candidates and potential drug targets.
Collapse
Affiliation(s)
- Vilbert Sikorski
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Pasi Karjalainen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Daria Blokhina
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Kati Oksaharju
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jahangir Khan
- Tampere Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland; (J.K.); (J.L.)
| | | | - Helena Rajala
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Satu Suihko
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Kari Teittinen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Annu Nummi
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Antti Nykänen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Arda Eskin
- Graduate School of Informatics, Department of Health Informatics, Middle East Technical University, 06800 Ankara, Turkey;
| | - Christoffer Stark
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Fausto Biancari
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
- Heart Center, Turku University Hospital and Department of Surgery, University of Turku, 20521 Turku, Finland
- Research Unit of Surgery, Anesthesiology and Critical Care, University of Oulu, 90014 Oulu, Finland
| | - Jan Kiss
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jarmo Simpanen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jussi Ropponen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Karl Lemström
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Kimmo Savinainen
- Clinical Biobank Tampere, Tampere University Hospital, 33520 Tampere, Finland;
| | - Maciej Lalowski
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland;
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Department of Biomedical Proteomics, 61-704 Poznan, Poland
| | - Markku Kaarne
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Mikko Jormalainen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Outi Elomaa
- Folkhälsan Research Center, 00250 Helsinki, Finland; (S.K.); (O.E.)
| | - Pertti Koivisto
- Chemistry Unit, Finnish Food Authority, 00790 Helsinki, Finland;
| | - Peter Raivio
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Pia Bäckström
- Helsinki Biobank, Hospital District of Helsinki and Uusimaa, 00029 Helsinki, Finland;
| | - Sebastian Dahlbacka
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Simo Syrjälä
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Tiina Vainikka
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Tommi Vähäsilta
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, College of Engineering, Koç University, 34450 Istanbul, Turkey;
- School of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, 50411 Tartu, Estonia;
| | - Eero Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Tatu Juvonen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
- Research Unit of Surgery, Anesthesiology and Critical Care, University of Oulu, 90014 Oulu, Finland
| | - Mika Laine
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jari Laurikka
- Tampere Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland; (J.K.); (J.L.)
| | - Antti Vento
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| |
Collapse
|
32
|
Atabekov TA, Batalov RE, Sazonova SI, Gusakova AM, Krivolapov SN, Khlynin MS, Popov SV. Role of stimulating growth factor 2 and galectin-3 in predicting the ventricular tachyarrhythmias in patients with ischemic cardiomyopathy. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To assess the role of stimulating growth factor 2 (ST-2) and galectin-3 in predicting the ventricular tachyarrhythmias (VTA) in patients with coronary artery disease (CAD) and left ventricular ejection fraction (LVEF) <35%.Material and methods. The study included 40 patients (men, 36 (90,0%); median age, 64,5 [57,5; 68,5] years) with CAD, NYHA class IIIII heart failure, LVEF <35%, indications for implantable cardioverter defibrillator (ICD) (primary prevention of sudden cardiac death). Prior to ICD implantation, patients were measured for serum ST-2 and galectin-3 levels. For 18 months, patients were assessed for arrhythmic events recorded in the ICD memory.Results. The 1st group consisted of 10 (25,0%) patients who had VTA episodes, arrested by antitachycardia pacing or shock during 18-month follow-up. The 2nd group consisted of 30 (75,0%) patients without VTA episodes. It was found that ST-2 concentration >22,48 ng/ml (p=0,02) and galectin-3 >10,95 ng/ml (p=0,009) corresponded to appropriate ICD discharge. Multivariate ROC analysis demonstrated that the only independent predictor of VTA was ST-2 elevation (odds ratio, 1,11; 95% CI, 1,01-1,21; p=0,023).Conclusion. An increase in the concentration of both ST-2 >22,48 ng/ml and galectin-3 >10,95 ng/ml had a high predictive value in assessing the VTA risk in patients with ischemic cardiomyopathy. In multivariate analysis, an increase in ST-2 >22,48 ng/ml was an independent predictor of VTA.
Collapse
Affiliation(s)
- T. A. Atabekov
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - R. E. Batalov
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - S. I. Sazonova
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - A. M. Gusakova
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - S. N. Krivolapov
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - M. S. Khlynin
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - S. V. Popov
- Cardiology Research Institute, Tomsk National Research Medical Center
| |
Collapse
|
33
|
Kim AJ, Ro H, Kim H, Ko KP, Chang JH, Lee HH, Chung W, Jung JY. Elevated levels of soluble ST2 but not galectin-3 are associated with increased risk of mortality in hemodialysis patients. Kidney Res Clin Pract 2021; 40:109-119. [PMID: 33706479 PMCID: PMC8041640 DOI: 10.23876/j.krcp.20.133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/01/2020] [Indexed: 11/07/2022] Open
Abstract
Background The soluble forms of suppression of tumorigenicity-2 (ST2) and galectin-3 have been proposed as novel biomarkers for cardiac fibrosis and heart failure, as well as predictors of cardiovascular events and mortality. However, there are limited data on the association between soluble ST2 and galectin-3 and clinical outcomes in patients with kidney failure on replacement therapy. To determine this, we examined the associations between soluble ST2 and galectin-3 and all-cause mortality and cardiovascular events in patients on hemodialysis. Methods This study included maintenance hemodialysis patients (over 18 years old) who consented to preserve their serum in the Biobank at our institution between March 2014 and March 2015. We used Cox proportional hazards regression analysis to evaluate the associations between soluble ST2, galectin-3 levels, and clinical outcomes. The primary outcome was all-cause mortality, the secondary outcome was cardiovascular disease, and patients were followed for both outcomes until March 2018. Results A total of 296 patients were analyzed in this study. The mean age was 57 ± 13 years, and 53.0% were male. Serum concentration of soluble ST2 was significantly associated with higher mortality, after adjustment for confounding factors, but was not associated with cardiovascular disease. Serum galectin-3 level was not independently associated with either outcome after adjustment. Conclusion Elevated soluble ST2 is independently associated with an increased risk of mortality, but not with cardiovascular disease, in patients on hemodialysis. Elevated galectin-3 was not associated with mortality or cardiovascular disease.
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
| | - Kwang-Pil Ko
- Department of Preventive Medicine, Gachon University College of Medicine, 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
|
34
|
Kalstad AA, Myhre PL, Laake K, Opstad TB, Tveit A, Solheim S, Arnesen H, Seljeflot I. Biomarkers of ageing and cardiac remodeling are associated with atrial fibrillation. SCAND CARDIOVASC J 2021; 55:213-219. [PMID: 33650449 DOI: 10.1080/14017431.2021.1889653] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objectives. Ageing is one of the strongest risk factors for atrial fibrillation (AF), and additional risk factors are also closely related to ageing. Remodeling is part of the pathophysiology of AF, and a possible common denominator of ageing and other AF risk factors. The aim of this study was to investigate any association between the presence of AF and the ageing biomarkers, leukocyte telomere length (LTL) and sirtuin-1 (SIRT-1), and the cardiac remodeling biomarkers Galectin-3 and sST2 in elderly myocardial infarction (MI) patients. Design. Patients were included after admission for MI. Diagnosis of AF was retrieved from medical records and classified as either history of AF before MI or new onset from admission to study inclusion. SIRT-1, sST2 and Galectin-3 were analyzed by ELISAs and LTL by qPCR. Results. In total, 299 patients were included, median age 75 years, 70.2% male. A history of AF was recorded in 38 patients and 30 patients experienced new onset AF. Higher levels of SIRT-1 were associated with lower risk of having a history of AF (OR = 0.46 (95% CI 0.26, 0.81), p = 0.007), whereas higher sST2 levels were associated with higher risk of AF (OR = 4.13 (95% CI 1.69, 10.13), p = 0.002). Results remained significant after adjustment for other AF risk factors. No significant associations with AF were found for Galectin-3 or LTL. None of the biomarkers associated with new onset AF. Conclusion. In elderly patients with MI, higher ST2 and lower SIRT-2 levels were associated with higher prevalence of AF, possibly reflecting both ageing and the remodeling phenomena in AF. Clinical trials registration: ClinicalTrials.gov (NCT01841944).
Collapse
Affiliation(s)
- Are A Kalstad
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Medicine, Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Kristian Laake
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Trine B Opstad
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arnljot Tveit
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Medical Research, Vestre Viken Hospital Trust, Baerum Hospital, Gjettum, Norway
| | - Svein Solheim
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Harald Arnesen
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Li M, Duan L, Cai Y, Hao B, Chen J, Li H, Liu H. Prognostic value of soluble suppression of tumorigenesis-2 (sST2) for cardiovascular events in coronary artery disease patients with and without diabetes mellitus. Cardiovasc Diabetol 2021; 20:49. [PMID: 33608010 PMCID: PMC7896409 DOI: 10.1186/s12933-021-01244-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background Soluble suppression of tumorigenesis-2 (sST2) is implicated in myocardial overload and has long been recognized as an inflammatory marker related to heart failure and acute coronary syndrome, but data on the prognostic value of sST2 in patients with coronary artery disease (CAD) remain limited. This study sought to investigate the prognostic value of sST2 in patients with established CAD and its predictive value in CAD patients with and without type 2 diabetes mellitus (T2DM). Methods A total of 3641 consecutive patients were included in this prospective cohort study. The primary end point was major adverse cardiovascular events (MACEs). The secondary end point was all-cause death. The association between sST2 and outcomes was investigated using multivariable Cox regression. Results During a median follow-up of 6.4 years, MACEs occurred in 775 patients, and 275 patients died. Multiple Cox regression models showed that a higher level of sST2 was an independent predictor of MACEs development (HR = 1.36, 95% CI 1.17–1.56, p < 0.001) and all-cause death (HR = 2.01, 95% CI 1.56–2.59, p < 0.001). The addition of sST2 to established risk factors significantly improved risk prediction of the composite outcome of MACEs and all-cause death (C-index, net reclassification index, and integrated discrimination improvement, all p < 0.05). In subgroup analysis depending on diabetes status, the diabetes group had a significantly higher level of sST2, which remained a significant predictor of MACEs and all-cause death in patients with and without T2DM in multivariable models. The area under the curve (AUC) of CAD patients with diabetes mellitus was significantly higher than that of those without T2DM. For MACEs, the AUC was 0.737 (patients with T2DM) vs 0.620 (patients without T2DM). For all-cause death, the AUC was 0.923 (patients with T2DM) vs 0.789 (patients without T2DM). Conclusions A higher level of sST2 is significantly associated with long-term MACEs and all-cause death in CAD patients with and without T2DM. sST2 has strong predictive value for cardiovascular adverse events in CAD patients with T2DM, and these results provide new evidence for the role of sST2.
Collapse
Affiliation(s)
- Man Li
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Lei Duan
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Yulun Cai
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Benchuan Hao
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Jianqiao Chen
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Huiying Li
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China. .,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, China.
| |
Collapse
|
36
|
Salem SS, Saleh NY, Soliman SE, Abo-Haded HM. On-admission plasma levels of BNP, MR-proADM, and cTnI in pediatric heart failure: contributions to diagnosis, prognosis, and outcome. Ir J Med Sci 2021; 191:263-270. [PMID: 33564973 DOI: 10.1007/s11845-021-02533-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/28/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The aim of this study is to evaluate the use of on-admission plasma levels of BNP, MR-proADM, and cTnI in diagnosing the clinical severity and progression of heart failure (HF) in children with CHD. Also, to correlate the levels of these biomarkers with the HF outcome (survival versus in-hospital mortality). RESULTS A prospective cohort study conducted in period from January 2017 to March 2018. All children presenting with HF had a Ross score assessment, echocardiography, and on-admission plasma level assay of BNP, MR-proADM, and cTnI. Patients were followed clinically throughout their hospital stay. The discriminatory power of on-admission measurement of each biomarker was determined using the receiver-operating characteristic (ROC). The results showed a significantly high on-admission plasma level of the 3 biomarkers among CHD cohort children than healthy controls (p < 0.001). Linear correlation was noted between the 3 biomarkers with Ross score, ejection fraction, and duration of hospital stay. Furthermore, significant association between on-admission level of the 3 biomarkers (BNP, MR-proADM, and cTnI) with patient's in-hospital mortality (p = 0.0003, Beta coefficient = 0.842; p = 0.0495, Beta coefficient = 0.183; and p < 0.001, Beta coefficient = 0.635, respectively), with on-admission BNP (cut of point 507.13) predicting in-hospital mortality, with 95.5% sensitivity, 88% specificity. CONCLUSIONS There is a high diagnostic value of measuring the on-admission levels of BNP, MR-proADM, and cTnI regarding the clinical severity and disease progression in the setting of pediatric heart failure, but the BNP level was more superior in prediction of the patients' outcome.
Collapse
Affiliation(s)
- Sherif S Salem
- Department of Pediatrics, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Nagwan Y Saleh
- Department of Pediatrics, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Shimaa E Soliman
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Hany M Abo-Haded
- Pediatric Cardiology Unit, Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
37
|
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: 6] [Impact Index Per Article: 1.5] [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
|
38
|
Galectin-3 and sST2 as Prognosticators for Heart Failure Requiring Extracorporeal Life Support: Jack n' Jill. Biomolecules 2021; 11:biom11020166. [PMID: 33513858 PMCID: PMC7911521 DOI: 10.3390/biom11020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 11/18/2022] Open
Abstract
Extracorporeal life support provides perfusion for patients with heart failure to allow time for recovery, function as a bridge for patients to heart transplantation, or serve as destination therapy for long term mechanical device support. Several biomarkers have been employed in attempt to predict these outcomes, but it remains to be determined which are suitable to guide clinical practice relevant to extracorporeal life support. Galectin-3 and soluble suppression of tumorigenicity-2 (sST2) are two of the more promising candidates with the greatest supporting evidence. In this review, we address the similarities and differences between galectin-3 and sST2 for prognostic prediction in adults and children with heart failure requiring extracorporeal life support and highlight the significant lack of progress in pediatric biomarker discovery and utilization.
Collapse
|
39
|
Avcı A, Somuncu MU, Can M, Akgul F. Could sST2 Predict Contrast-Induced Nephropathy in ST-Segment Elevation Myocardial Infarction? Int J Gen Med 2020; 13:1297-1304. [PMID: 33273849 PMCID: PMC7708263 DOI: 10.2147/ijgm.s287834] [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: 10/21/2020] [Accepted: 11/12/2020] [Indexed: 01/09/2023] Open
Abstract
Background and Aim One of the most worrying complications of primary percutaneous coronary interventions is contrast-induced nephropathy (CIN) that is associated with increased mortality and morbidity in myocardial infarction. In this study, we questioned whether soluble suppression of tumorigenesis-2 (sST2), which has thought to play a role in inflammatory processes, cardiac remodeling, and fibrosis could give an idea about the development of CIN in ST-elevation myocardial infarction (STEMI) patients. Patients and Methods This study is a cross-sectional observational study and includes 357 consecutive STEMI patients. Demographic features, medical history, laboratory parameters, and procedural characteristics were compared according to CIN's development. The multivariate logistic regression analysis was selected to detect independent risk factors of CIN. Results In the study, 81 patients (22.7%) who developed CIN were identified. The concentration of sST2 in CIN (+) group was higher than that of CIN (-) group (40.6±21.0 ng/mL vs 31.5±13.0 ng/L, p<0.001). Independent predictors of CIN development were diabetes mellitus (OR, 2.059; 95% CI, 1.093-3.879; p=0.025), eGFR (OR, 0.983; 95% CI, 0.972-0.995; p=0.006), lower systolic blood pressure (OR, 0.976; 95% CI, 0.960-0.993; p=0.006), total procedure time (OR, 1.030; 95% CI, 1.011-1.049; p=0.002), and sST2 (OR, 1.101; 95% CI; 1.046-1.160; p<0.001). Besides, the risk of developing CIN in the high sST2 group is 3.06 times higher than the low group sST2 group regardless of other risk factors. Conclusion sST2 levels on admission in STEMI patients are useful in predicting CIN development.
Collapse
Affiliation(s)
- Ahmet Avcı
- Department of Cardiology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Mustafa Umut Somuncu
- Department of Cardiology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Murat Can
- Department of Biochemistry, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Ferit Akgul
- Department of Cardiology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
40
|
Hansen C, Sastre C, Wolcott Z, Bevers MB, Kimberly WT. Time-dependent, dynamic prediction of fatty acid-binding protein 4, Galectin-3, and soluble ST2 measurement with poor outcome after acute stroke. Int J Stroke 2020; 16:660-668. [PMID: 33167787 DOI: 10.1177/1747493020971166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Time-dependent change in the level of biomarkers after stroke is not well understood. We sought to compare fatty acid-binding protein 4 (FABP4), Galectin-3, and soluble ST2 to ascertain for a change in prediction of outcome at admission and 48 h later. METHODS Plasma FABP4, Galectin-3, and soluble ST2 were measured in biospecimens from acute stroke patients at the time of admission (n = 383) and 48 h later (n = 244). Functional outcome was assessed at 90 days using the modified Rankin Scale and dichotomized into good (modified Rankin Scale 0-2) and poor outcome (modified Rankin Scale 3-6). RESULTS On admission, elevated levels of each biomarker predicted poor outcome (FABP4: OR 1.92, 95% CI 1.42-2.59, P < 0.0001; Galectin-3: OR 1.85, 95% CI 1.42-2.40, P < 0.0001; soluble ST2: OR 1.55, 95% CI 1.22-1.97, P < 0.0001) and death (FABP4: OR 2.45; 95% CI 1.51-3.98; P < 0.0001; Galectin-3: OR 2.12; 95% CI 1.50-3.30; P < 0.0001; soluble ST2: OR 2.17; 95% CI 1.58-2.99; P < 0.0001). At 48 h, soluble ST2 predicted poor outcome (OR 2.62, 95% CI 1.77-3.88, P < 0.0001) and mortality (OR 3.36, 95% CI 2.06-5.48, P < 0.0001), and Galectin-3 predicted mortality only (OR 1.81, 95% CI 1.05-3.10, P = 0.033). FABP4 measured at 48 h was not predictive of outcome or death. Associations of Galectin-3 and soluble ST2 with outcome or mortality were independent of age, sex, and NIHSS, whereas those with FABP4 were not. CONCLUSIONS Galectin-3 performed better when measured on admission, whereas soluble ST2 was predictive at admission and better at 48 h after stroke. The time-dependent differences may reflect the evolving role of these pathways after acute stroke.
Collapse
Affiliation(s)
- Christina Hansen
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina Sastre
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zoe Wolcott
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew B Bevers
- Divisions of Stroke, Cerebrovascular and Critical Care Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - W Taylor Kimberly
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
41
|
IL-33 induces type-2-cytokine phenotype but exacerbates cardiac remodeling post-myocardial infarction with eosinophil recruitment, worsened systolic dysfunction, and ventricular wall rupture. Clin Sci (Lond) 2020; 134:1191-1218. [PMID: 32432676 DOI: 10.1042/cs20200402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/02/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is the leading cause of mortality worldwide. Interleukin (IL)-33 (IL-33) is a cytokine present in most cardiac cells and is secreted on necrosis where it acts as a functional ligand for the ST2 receptor. Although IL-33/ST2 axis is protective against various forms of cardiovascular diseases, some studies suggest potential detrimental roles for IL-33 signaling. The aim of the present study was to examine the effect of IL-33 administration on cardiac function post-MI in mice. MI was induced by coronary artery ligation. Mice were treated with IL-33 (1 μg/day) or vehicle for 4 and 7 days. Functional and molecular changes of the left ventricle (LV) were assessed. Single cell suspensions were obtained from bone marrow, heart, spleen, and peripheral blood to assess the immune cells using flow cytometry at 1, 3, and 7 days post-MI in IL-33 or vehicle-treated animals. The results of the present study suggest that IL-33 is effective in activating a type 2 cytokine milieu in the damaged heart, consistent with reduced early inflammatory and pro-fibrotic response. However, IL-33 administration was associated with worsened cardiac function and adverse cardiac remodeling in the MI mouse model. IL-33 administration increased infarct size, LV hypertrophy, cardiomyocyte death, and overall mortality rate due to cardiac rupture. Moreover, IL-33-treated MI mice displayed a significant myocardial eosinophil infiltration at 7 days post-MI when compared with vehicle-treated MI mice. The present study reveals that although IL-33 administration is associated with a reparative phenotype following MI, it worsens cardiac remodeling and promotes heart failure.
Collapse
|
42
|
Shah NN, Ayyadurai P, Saad M, Kosmas CE, Dogar MU, Patel U, Vittorio TJ. Galactin-3 and soluble ST2 as complementary tools to cardiac MRI for sudden cardiac death risk stratification in heart failure: A review. JRSM Cardiovasc Dis 2020; 9:2048004020957840. [PMID: 33088496 DOI: 10.1177/2048004020957840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 11/17/2022] Open
Abstract
Heart failure (HF) is recognized as one of the leading causes of morbidity and mortality worldwide. Every year about 500,000 new cases of HF are diagnosed in the United States. The predominant etiology of death in HF patients include sudden cardiac death (SCD) and pump failure. Prediction of mode of death may help in devising management decisions. In patients with HF, the presence of myocardial fibrosis has been a known risk factor for SCD and thus it could be used as a criterion in risk stratification for SCD. However, the underlying pathophysiology of SCD is uncertain and controversial, which makes it necessary to develop newer tools to enhance SCD risk stratification. The newer tools should be innovative enough either to complement or to replace the currently available tools. In this scoping review, we highlighted the utilization of novel biomarkers galectin-3 (gal-3) and soluble ST2 (sST2) and discussed that how they might complement currently available tools such as, cardiac MRI (CMR) for SCD risk stratification in HF patients.
Collapse
Affiliation(s)
- Niel N Shah
- Department of Medicine, BronxCare Health System/BronxCare Hospital Center, Bronx, NY, USA
| | - Puvanalingam Ayyadurai
- Department of Medicine, BronxCare Health System/BronxCare Hospital Center, Bronx, NY, USA
| | - Muhammad Saad
- Department of Medicine, BronxCare Health System/BronxCare Hospital Center, Bronx, NY, USA
| | - Constantine E Kosmas
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Muhammad U Dogar
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Upen Patel
- Department of Medicine, Nassau University Medical Center, East Meadow, NY, USA
| | - Timothy J Vittorio
- Department of Medicine, BronxCare Health System/BronxCare Hospital Center, Bronx, NY, USA
| |
Collapse
|
43
|
Liu N, Hang T, Gao X, Yang W, Kong W, Lou Q, Yang J. The association between soluble suppression of tumorigenicity-2 and long-term prognosis in patients with coronary artery disease: A meta-analysis. PLoS One 2020; 15:e0238775. [PMID: 32886697 PMCID: PMC7473587 DOI: 10.1371/journal.pone.0238775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Findings regarding the prognostic value of soluble suppression of tumorigenecity-2 (sST2) in patients with coronary artery disease (CAD) remain inconsistent. Therefore, we conducted this meta-analysis to investigate the long-term prognostic value of sST2 in patients with CAD. METHODS A comprehensive literature search was conducted across the PubMed, Embase, and Cochrane Library databases up to June 3, 2020. The primary outcome was major adverse cardiac events (MACEs). The secondary outcomes were all-cause mortality, cardiovascular (CV) death, heart failure (HF), and myocardial infarction (MI). Pooled estimations and 95% confidence intervals (CIs) were assessed using a random-effects model. RESULTS Twenty-two articles that enrolled a total of 17,432 patients with CAD were included in the final analysis. CAD patients in the highest categories of baseline sST2 had a significantly higher risk of MACEs (HR: 1.42, 95% CI: 1.09-1.76), all-cause mortality (HR: 2.00, 95% CI: 1.54-2.46), and CV death (HR: 1.42, 95% CI: 1.15-1.68), HF (HR: 2.41, 95% CI: 1.87-2.94), but not that of MI (HR: 1.15, 95% CI: -0.73-3.04), than those in the lowest categories. These results were consistent when baseline sST2 was presented as continuous values in one unit increments. Moreover, subgroup analysis showed that elevated baseline sST2 levels increased the long-term risk of MACEs in the acute coronary syndrome (ACS) population (HR: 1.74, 95% CI: 1.39-2.09) but only showed a trend toward higher risk of MACEs in the non-ACS population (HR: 1.09, 95% CI: 0.87-1.30). CONCLUSIONS The findings suggest that a higher concentration of baseline sST2 is associated with a higher risk of MACEs, all-cause mortality, CV death, and HF in patients with CAD. Elevated sST2 levels could significantly predict future MACEs in the ACS population but not in the non-ACS population.
Collapse
Affiliation(s)
- Niannian Liu
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Hang
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Gao
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxue Yang
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjie Kong
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiaozhen Lou
- Department of Cardiology, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiming Yang
- Department of Cardiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
Banaszkiewicz M, Pietrasik A, Darocha S, Piłka M, Florczyk M, Dobosiewicz A, Kędzierski P, Pędzich-Placha E, Kochman J, Opolski G, Torbicki A, Kurzyna M. Soluble ST2 protein as a new biomarker in patients with precapillary pulmonary hypertension. Arch Med Sci 2020; 20:1442-1451. [PMID: 39649252 PMCID: PMC11623185 DOI: 10.5114/aoms.2020.98635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/25/2020] [Indexed: 12/10/2024] Open
Abstract
Introduction Non-invasive tests that may improve clinical evaluation of pulmonary hypertension (PH) are needed. The purpose of this study was to assess the role of soluble ST2 (sST2) in patients with PH. Material and methods A total of 57 patients with chronic thromboembolic PH and 43 patients with idiopathic arterial PH were enrolled in this study. All patients were evaluated for World Health Organization (WHO) functional class (FC), N-terminal prohormone B-type natriuretic peptide (NT-proBNP), troponin T (TnT), and hemodynamics. Plasma sST2 was assessed by an immunofluorescent in vitro diagnostic assay. All patients were followed up from the date of blood sampling. The endpoint was all-cause death. Results The median sST2 concentration was 32.8 ng/ml (IQR: 21.6-48.5 ng/ml) in the whole study population. Significant differences were found between median sST2 in successive WHO FCs (FC II vs. FC III, p = 0.002; FC III vs. FC IV, p = 0.12; FC II vs. FC IV, p = 0.008). Significant correlations were found between sST2 and hemodynamic parameters: mean right atrial pressure (r = 0.56; p < 0.05), mean pulmonary artery pressure (r = 0.25; p < 0.05), cardiac index (r = -0.40; p < 0.05), pulmonary vascular resistance (r = 0.41; p < 0.05), and between sST2 and WHO FC (r = 0.36; p < 0.05), NT-proBNP (r = 0.55; p < 0.05), and TnT (r = 0.44; p < 0.05). sST2 concentration above the median was associated with worse clinical prognosis (p = 0.02, Kaplan-Meier). Conclusions sST2 seems to be a marker of poor clinical prognosis in patients with PH.
Collapse
Affiliation(s)
- Marta Banaszkiewicz
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Arkadiusz Pietrasik
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Szymon Darocha
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Michał Piłka
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Michał Florczyk
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Anna Dobosiewicz
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Piotr Kędzierski
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Ewa Pędzich-Placha
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Janusz Kochman
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Opolski
- First Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Adam Torbicki
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| | - Marcin Kurzyna
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Poland
| |
Collapse
|
45
|
Tian X, Guo Y, Wang X, Pei L, Wang X, Wu J, Sun S, Li Y, Ning M, Buonanno FS, Xu Y, Song B. Serum soluble ST2 is a potential long-term prognostic biomarker for transient ischaemic attack and ischaemic stroke. Eur J Neurol 2020; 27:2202-2208. [PMID: 32593220 DOI: 10.1111/ene.14419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Soluble ST2 (sST2) is a promising biomarker in inflammation, atherosclerosis and cardiovascular diseases. We investigated the association between serum sST2 and poor outcome in patients with transient ischaemic attack (TIA)/ischaemic stroke. METHODS Patients within 24 h after onset and with measured serum sST2 were prospectively enrolled in this study. Poor outcome was a combination of a new stroke event (ischaemic or haemorrhagic) and all-cause death within 90 days and 1 year. The associations of serum sST2 with poor outcome were analysed by Cox proportional hazards. RESULTS Among the 430 patients included, the median (interquartile range) sST2 was 17.72 (9.31-28.84) ng/mL. A total of 19 (4.4%) and 38 (8.8%) patients experienced poor outcome within 90 days and 1 year, respectively. Compared with the lowest sST2 tertile, hazard ratios (HRs) [95% confidence intervals (CI)] for the highest tertile were 5.14 (1.43-18.51) for poor outcome within 90 days and 3.00 (1.29-6.97) at 1 year after multivariate adjustments. Adding sST2 to a prediction model significantly improved risk stratification of poor outcome in TIA/ischaemic stroke, as observed by the continuous net reclassification improvement of 60.98% (95% CI, 15.37-106.6%, P = 0.009) and integrated discrimination improvement of 2.63% (95% CI, 0.08-5.18%, P = 0.043) at 90 days and the continuous net reclassification improvement of 41.68% (95% CI, 8.74-74.61%, P = 0.013) at 1 year. CONCLUSIONS Increased serum sST2 levels in TIA/ischaemic stroke were associated with increased risks of poor outcome within 90 days and 1 year, suggesting that serum sST2 may be a potential long-term prognostic biomarker for TIA/ischaemic stroke.
Collapse
Affiliation(s)
- X Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - Y Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - X Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - L Pei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - X Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - J Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - S Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - Y Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - M Ning
- Clinical Proteomics Research Centre and Cardio-Neurology Clinic, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - F S Buonanno
- Clinical Proteomics Research Centre and Cardio-Neurology Clinic, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Y Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| | - B Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Cerebrovascular Diseases, Zhengzhou, China
| |
Collapse
|
46
|
Nah EH, Cho S, Kim S, Cho HI. Reference interval and the role of soluble suppression of tumorigenicity 2 (sST2) in subclinical cardiac dysfunction at health checkups. J Clin Lab Anal 2020; 34:e23461. [PMID: 32638437 PMCID: PMC7676181 DOI: 10.1002/jcla.23461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 01/02/2023] Open
Abstract
Background Soluble ST2 (sST2) is known to predict adverse outcomes and death in individuals with established heart failure. However, the role of sST2 testing in the general population has not been established. The aims of this study were to determine the reference interval (RI) and the clinical utility of sST2 in subclinical cardiac dysfunction in general population. Methods This cross‐sectional study consecutively selected 41,806 general subjects at health checkups who underwent echocardiography and sST2 testing at 16 health promotion centers in 13 Korean cities. The reference subjects were obtained among those with normal findings in echocardiography. Sex‐specific RIs were established according to the CLSI C28‐A3 guidelines. sST2 was measured using immunoassay with the Presage ST2 assay (Critical Diagnostics). Results In the general subjects, age, sex, BMI, systolic blood pressure, blood glucose, creatinine, liver function, and triglycerides were associated with the sST2 levels. The RI for sST2 was higher in males (≤49.6 ng/mL, 95% CI = 48.5‐51.5) than in females (≤44.5 ng/mL, 95% CI = 43.5‐45.6) and higher in subjects aged < 40 years than ≥ 40 years in both sexes. The sST2 levels were 29.1 ± 10.7 (mean ± SD) and 29.1 ± 14.4 ng/mL in the groups with normal cardiac function and subclinical cardiac dysfunction, respectively. The sST2 level was not associated with subclinical cardiac dysfunction (odd ratio = 1.002, P = .13). Conclusions RIs obtained from a large and echocardiography‐proven healthy community‐based sample are presented. Subclinical cardiac dysfunction was associated with older age, male sex, and metabolic factors but not with the sST2 level.
Collapse
Affiliation(s)
- Eun-Hee Nah
- Department of Laboratory Medicine, Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Seon Cho
- Department of Laboratory Medicine, Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Suyoung Kim
- Department of Laboratory Medicine, Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Han-Ik Cho
- MEDIcheck LAB, Korea Association of Health Promotion, Cheongju, Korea
| |
Collapse
|
47
|
Giurgea GA, Zlabinger K, Gugerell A, Lukovic D, Syeda B, Mandic L, Pavo N, Mester-Tonczar J, Traxler-Weidenauer D, Spannbauer A, Kastner N, Müller C, Anvari A, Bergler-Klein J, Gyöngyösi M. Multimarker Approach to Identify Patients with Coronary Artery Disease at High Risk for Subsequent Cardiac Adverse Events: The Multi-Biomarker Study. Biomolecules 2020; 10:biom10060909. [PMID: 32549327 PMCID: PMC7356937 DOI: 10.3390/biom10060909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
In our prospective non-randomized, single-center cohort study (n = 161), we have evaluated a multimarker approach including S100 calcium binding protein A12 (S100A1), interleukin 1 like-receptor-4 (IL1R4), adrenomedullin, copeptin, neutrophil gelatinase-associated lipocalin (NGAL), soluble urokinase plasminogen activator receptor (suPAR), and ischemia modified albumin (IMA) in prediction of subsequent cardiac adverse events (AE) during 1-year follow-up in patients with coronary artery disease. The primary endpoint was to assess the combined discriminatory predictive value of the selected 7 biomarkers in prediction of AE (myocardial infarction, coronary revascularization, death, stroke, and hospitalization) by canonical discriminant function analysis. The main secondary endpoints were the levels of the 7 biomarkers in the groups with/without AE; comparison of the calculated discriminant score of the biomarkers with traditional logistic regression and C-statistics. The canonical correlation coefficient was 0.642, with a Wilk’s lambda value of 0.78 and p < 0.001. By using the calculated discriminant equation with the weighted mean discriminant score (centroid), the sensitivity and specificity of our model were 79.4% and 74.3% in prediction of AE. These values were higher than that of the calculated C-statistics if traditional risk factors with/without biomarkers were used for AE prediction. In conclusion, canonical discriminant analysis of the multimarker approach is able to define the risk threshold at the individual patient level for personalized medicine.
Collapse
Affiliation(s)
- Georgiana-Aura Giurgea
- Department of Angiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Katrin Zlabinger
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Alfred Gugerell
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Dominika Lukovic
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Bonni Syeda
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Ljubica Mandic
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Noemi Pavo
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Julia Mester-Tonczar
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Denise Traxler-Weidenauer
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Andreas Spannbauer
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Nina Kastner
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Claudia Müller
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Anahit Anvari
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Jutta Bergler-Klein
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
| | - Mariann Gyöngyösi
- Department of Cardiology, Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (K.Z.); (A.G.); (D.L.); (B.S.); (L.M.); (N.P.); (J.M.-T.); (D.T.-W.); (A.S.); (N.K.); (C.M.); (A.A.); (J.B.-K.)
- Correspondence: ; Tel.: +43-1-40400-46140
| |
Collapse
|
48
|
Firouzabadi N, Dashti M, Dehshahri A, Bahramali E. Biomarkers of IL-33 and sST2 and Lack of Association with Carvedilol Therapy in Heart Failure. Clin Pharmacol 2020; 12:53-58. [PMID: 32607003 PMCID: PMC7305854 DOI: 10.2147/cpaa.s256290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/29/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE The IL-33/ST2 pathway plays a fundamental role in the cardiovascular system and can be considered as a new therapeutic strategy for the treatment or prevention of cardiovascular diseases. ST2, as an interleukin (IL)-1 receptor family member, has transmembrane (ST2L) and soluble (sST2) isoforms. sST2 neutralizes IL-33 and thereby inhibits the cardioprotective role of IL-33/ST2L signaling pathway. Increase in sST2 level is associated with weak cardiac output and can be a predictor of mortality in heart failure (HF). Thereby, we hypothesized that there may be a relationship between the cardioprotective effects of carvedilol and sST2 and IL-3 in HF patients. METHODS sST2 and IL-33 were measured in serum of 66 individuals; 22 healthy volunteers and 44 suffering from HF; among whom 25 patients received carvedilol and the other 19 patients did not receive any β-blockers. RESULTS Lack of association between serum levels of IL-33 and sST2 was observed between HF patients and healthy individuals (2.4466 ± 0.69 vs 2.6748 ± 0.33 and 3416.6 ± 1089.1 vs 2971.6 ± 792.5, respectively). Our results indicated no significant difference between sST2 and IL-33 levels in HF patients who did not receive beta-blockers and patients receiving carvedilol (P=0.59 and P=0.97). CONCLUSION Our results showed a lack of association between serum levels of IL-33 and sST2 and HF. Moreover, the results do not confirm the cardioprotective mechanism of carvedilol by means of IL-33/sST2 pathway.
Collapse
Affiliation(s)
- Negar Firouzabadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Dashti
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Bahramali
- Digestive Disease Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Berezin AE, Berezin AA. Circulating Cardiac Biomarkers in Diabetes Mellitus: A New Dawn for Risk Stratification-A Narrative Review. Diabetes Ther 2020; 11:1271-1291. [PMID: 32430864 PMCID: PMC7261294 DOI: 10.1007/s13300-020-00835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of this narrative review is to update the current knowledge on the differential choice of circulating cardiac biomarkers in patients with prediabetes and established type 2 diabetes mellitus (T2DM). There are numerous circulating biomarkers with unconfirmed abilities to predict clinical outcomes in pre-DM and DM individuals; the prognostication ability of the cardiac biomarkers reported here has been established, and they are still being studied. The conventional cardiac biomarkers, such as natriuretic peptides (NPs), soluble suppressor tumorigenisity-2, high-sensitivity circulating cardiac troponins and galectin-3, were useful to ascertain cardiovascular (CV) risk. Each cardiac biomarker has its strengths and weaknesses that affect the price of usage, specificity, sensitivity, predictive value and superiority in face-to-face comparisons. Additionally, there have been confusing reports regarding their abilities to be predictably relevant among patients without known CV disease. The large spectrum of promising cardiac biomarkers (growth/differential factor-15, heart-type fatty acid-binding protein, cardiotrophin-1, carboxy-terminal telopeptide of collagen type 1, apelin and non-coding RNAs) is discussed in the context of predicting CV diseases and events in patients with known prediabetes and T2DM. Various reasons have been critically discussed related to the variable findings regarding biomarker-based prediction of CV risk among patients with metabolic disease. It was found that NPs and hs-cTnT are still the most important tools that have an affordable price as well as high sensitivity and specificity to predict clinical outcomes among patients with pre-DM and DM in routine clinical practice, but other circulating biomarkers need to be carefully investigated in large trials in the future.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, Ministry of Health of Ukraine, State Medical University, Zaporozhye, 69035, Ukraine.
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, 69096, Ukraine
| |
Collapse
|
50
|
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
|