1
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
2
|
Mas-Fontao S, Tarín N, Cristóbal C, Soto-Catalán M, Pello A, Aceña A, Lumpuy-Castillo J, Garces C, Gomez-Guerrero C, Gutiérrez-Landaluce C, Blanco-Colio LM, Martín-Ventura JL, Huelmos A, Alonso J, López Bescós L, Moreno JA, Mahíllo-Fernández I, Lorenzo Ó, González-Casaus ML, Egido J, Tuñón J. Elevated plasma levels of TNF-R1 predict the development of acute ischemic events in coronary patients with diabetes. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024:S0214-9168(24)00084-6. [PMID: 39343690 DOI: 10.1016/j.arteri.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES To examine the relationship between inflammatory biomarkers and the occurrence of cardiovascular events in patients with type 2 diabetes mellitus (DM2) and stable coronary artery disease. METHODS A total of 964 patients with stable coronary artery disease were included. Plasma levels of inflammatory markers, including tumour necrosis factor receptors 1 and 2 (TNF-R1 and TNF-R2), growth differentiation factor-15 (GDF-15), soluble suppression of tumorigenicity 2 (sST2), and high-sensitivity C-reactive protein (hsCRP) were measured. The primary endpoint was the development of acute ischaemic events (any type of acute coronary syndrome, stroke, or transient ischaemic attack). RESULTS There were 232 diabetic patients and 732 non-diabetic patients. Patients with coronary artery disease and DM2 (232, 24%) had higher levels of TNF-R1, TNF-R2, GDF-15, sST2 (P<.001), and hsCRP compared to patients without DM2, indicating a higher inflammatory state. After a median follow-up of 5.39 (2.81-6.92) years, patients with DM2 more frequently developed the primary endpoint (15.9% vs 10.8%; P=.035). Plasma levels of TNF-R1 were independent predictors of the primary endpoint in patients with DM2, along with male gender, triglyceride levels, and the absence of treatment with angiotensin-converting enzyme inhibitors. None of these inflammatory markers predicted the development of this event in non-diabetic patients. CONCLUSIONS Patients with stable coronary artery disease and DM2 exhibit elevated levels of the proinflammatory markers TNF-R1, TNF-R2, GDF-15, and sST2. Moreover, TNF-R1 is an independent predictor of acute ischaemic events only in diabetic patients.
Collapse
Affiliation(s)
- Sebastián Mas-Fontao
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España; Faculty of Medicine and Biomedicine, Universidad Alfonso X el Sabio (UAX), Madrid, España
| | - Nieves Tarín
- Department of Cardiology, Hospital Universitario de Móstoles, Móstoles, Madrid, España; Faculty of Medicine, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| | - Carmen Cristóbal
- Faculty of Medicine, Universidad Rey Juan Carlos, Alcorcón, Madrid, España; Department of Cardiology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Madrid, España
| | - Manuel Soto-Catalán
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España
| | - Ana Pello
- Department of Cardiology, IIS-Fundación Jiménez Díaz, Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España
| | - Alvaro Aceña
- Department of Cardiology, IIS-Fundación Jiménez Díaz, Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España
| | - Jairo Lumpuy-Castillo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España
| | - Carmen Garces
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Carmen Gomez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España
| | | | - Luis M Blanco-Colio
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; CIBERCV, Madrid, España
| | - José Luis Martín-Ventura
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España; CIBERCV, Madrid, España
| | - Ana Huelmos
- Department of Cardiology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, España
| | - Joaquín Alonso
- Faculty of Medicine, Universidad Rey Juan Carlos, Alcorcón, Madrid, España; Department of Cardiology, Hospital de Getafe, Getafe, Madrid, España
| | | | - Juan A Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, España; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Córdoba, España
| | - Ignacio Mahíllo-Fernández
- Department of Epidemiology and Biostatistics Research Unit, IIS-Fundación Jiménez Díaz, Madrid, España
| | - Óscar Lorenzo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España
| | | | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España.
| | - José Tuñón
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, España; Department of Cardiology, IIS-Fundación Jiménez Díaz, Madrid, España; Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, España; CIBERCV, Madrid, España.
| |
Collapse
|
3
|
Matyar S, Açıkalın Akpınar A, Dişel NR, Avci A, Çağlayan ÇE, Yıldırım A, Akpınar O. Prognostic value of sst2 in long-term mortality in acute heart failure. Acta Cardiol 2024:1-11. [PMID: 39317343 DOI: 10.1080/00015385.2024.2406683] [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: 06/13/2024] [Revised: 07/16/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND The use of biochemical markers in ADHF is considered valuable both in the diagnosis and treatment of diseases and in follow-up. This study aimed to investigate the prognostic power of serum sST2 and NT-proBNP levels in predicting long-term mortality in patients with ADHF using serial measurement. METHODS A total of 122 patients with ADHF were included in this prospective study. Venous blood samples were taken from the patients at the time of first admission to the emergency department and 48 h after hospitalisation. Serial measurements were performed using the same blood samples to determine NT-proBNP and sST2 levels. RESULTS The 1st time sST2 value was found to be significantly higher in the deceased group than in the living group, and this increase was found to be statistically significant (p < 0.001). The cut-off value for the 1st time value of sST2 was > 56.79 ng/mL, with 91.2% sensitivity and 79.5% specificity (area under the curve (AUC): 0.902, 95% confidence interval (CI): 0.835-0.948, p < 0.001). The cut-off value for the 2nd time sST2 value was > 38.91 ng/mL, with 97.1% sensitivity and 81.8% specificity (AUC: 0.932, 95% CI: 0.872-0.970, p < 0.001). CONCLUSION In our study, sST2 gained value as a marker that should be included in panels with multiple markers. It seems more appropriate to recommend the serial measurement of sST2 in heart failure. LIMITATIONS OF OUR STUDY The sample size is relatively small and there is no standard in timing and numbers in serial measurements.
Collapse
Affiliation(s)
- Selcuk Matyar
- Central Laboratory, Department of Biochemistry, University of Health Sciences, Adana City Research and Training Hospital, Adana, Turkey
| | - Ayça Açıkalın Akpınar
- Department of Emergency Medicine, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Nezihat Rana Dişel
- Department of Emergency Medicine, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Akkan Avci
- Department of Emergency Medicine, University of Health Sciences, Adana City Research and Training Hospital, Adana, Turkey
| | - Çağlar Emre Çağlayan
- Department of Cardiology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Abdullah Yıldırım
- Department of Cardiology, University of Health Sciences, Adana City Research and Training Hospital, Adana, Turkey
| | - Onur Akpınar
- Department of Cardiology, Near East University Faculty of Medicine, Nicosia, Cyprus
| |
Collapse
|
4
|
Paterson MA, Pilbrow AP, Frampton CM, Cameron VA, Troughton RW, Pemberton CJ, Lund M, Devlin GP, Richards AM, Doughty RN, Palmer BR. Plasma soluble fms-like tyrosine kinase-1, placental growth factor, and vascular endothelial growth factor system gene variants as predictors of survival in heart failure. Eur J Heart Fail 2024; 26:1804-1813. [PMID: 38980212 DOI: 10.1002/ejhf.3368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/09/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024] Open
Abstract
AIMS Soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PlGF), components of the vascular endothelial growth factor (VEGF) system, play key roles in angiogenesis. Reports of elevated plasma levels of sFlt-1 and PlGF in coronary heart disease and heart failure (HF) led us to investigate their utility, and VEGF system gene single nucleotide polymorphisms (SNPs), as prognostic biomarkers in HF. METHODS AND RESULTS ELISA assays for sFlt-1, PlGF and N-terminal pro-B-type natriuretic peptide (NT-proBNP) were performed on baseline plasma samples from the PEOPLE cohort (n = 890), a study of outcomes among patients after an episode of acute decompensated HF. Eight SNPs potentially associated with sFlt-1 or PlGF levels were genotyped. sFlt-1 and PlGF were assayed in 201 subjects from the Canterbury Healthy Volunteers Study (CHVS) matched to PEOPLE participants. All-cause death was the major endpoint for clinical outcome considered. In PEOPLE participants, mean plasma levels for both sFlt-1 (125 ± 2.01 pg/ml) and PlGF (17.5 ± 0.21 pg/ml) were higher (both p < 0.044) than in the CHVS cohort (81.2 ± 1.31 pg/ml and 15.5 ± 0.32 pg/ml, respectively). sFlt-1 was higher in HF with reduced ejection fraction compared to HF with preserved ejection fraction (p = 0.005). The PGF gene SNP rs2268616 was univariately associated with death (p = 0.016), and was also associated with PlGF levels, as was rs2268614 genotype. Cox proportional hazards modelling (n = 695, 246 deaths) showed plasma sFlt-1, but not PlGF, predicted survival (hazard ratio 6.44, 95% confidence interval 2.57-16.1; p < 0.001) in PEOPLE, independent of age, NT-proBNP, ischaemic aetiology, diabetic status and beta-blocker therapy. CONCLUSIONS Plasma sFlt-1 concentrations have potential as an independent predictor of survival and may be complementary to established prognostic biomarkers in HF.
Collapse
Affiliation(s)
- Melinda A Paterson
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Anna P Pilbrow
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris M Frampton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Vicky A Cameron
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Richard W Troughton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris J Pemberton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Mayanna Lund
- Cardiology Department, Middlemore Hospital, Auckland, New Zealand
| | - Gerard P Devlin
- Department of Cardiology, Waikato District Health Board, Hamilton, New Zealand
| | - A Mark Richards
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Robert N Doughty
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Barry R Palmer
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- School of Health Sciences, College of Health, Massey University, Wellington, New Zealand
| |
Collapse
|
5
|
Wu L, Zhu X, Luo C, Zhao Y, Pan S, Shi K, Chen Y, Qiu J, Shen Z, Guo J, Jie W. Mechanistic role of RND3-regulated IL33/ST2 signaling on cardiomyocyte senescence. Life Sci 2024; 348:122701. [PMID: 38724005 DOI: 10.1016/j.lfs.2024.122701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/23/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Hyperinflammatory responses are pivotal in the cardiomyocyte senescence pathophysiology, with IL33 serving as a crucial pro-inflammatory mediator. Our previous findings highlighted RND3's suppressive effect on IL33 expression. This study aims to explore the role of RND3 in IL33/ST2 signaling activation and in cardiomyocyte senescence. Intramyocardial injection of exogenous IL33 reduces the ejection fraction and fractional shortening of rats, inducing the appearance of senescence-associated secretory phenotype (SASP) in myocardial tissues. Recombinant IL33 treatment of AC16 cardiomyocytes significantly upregulated expression of SASP factors like IL1α, IL6, and MCP1, and increased the p-p65/p65 ratio and proportions of SA-β-gal and γH2AX-positive cells. NF-κB inhibitor pyrrolidinedithiocarbamate ammonium (PDTC) and ST2 antibody astegolimab treatments mitigated above effects. RND3 gene knockout H9C2 cardiomyocytes using CRISPR/Cas9 technology upregulated IL33, ST2L, IL1α, IL6, and MCP1 levels, decreased sST2 levels, and increased SA-β-gal and γH2AX-positive cells. A highly possibility of binding between RND3 and IL33 proteins was showed by molecular docking and co-immunoprecipitation, and loss of RND3 attenuated ubiquitination mediated degradation of IL33; what's more, a panel of ubiquitination regulatory genes closely related to RND3 were screened using qPCR array. In contrast, RND3 overexpression in rats by injection of AAV9-CMV-RND3 particles inhibited IL33, ST2L, IL1α, IL6, and MCP1 expression in cardiac tissues, decreased serum IL33 levels, and increased sST2 levels. These results suggest that RND3 expression in cardiomyocytes modulates cell senescence by inhibiting the IL33/ST2/NF-κB signaling pathway, underscoring its potential as a therapeutic target in cardiovascular senescence.
Collapse
Affiliation(s)
- Linxu Wu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China; Public Research Center of Hainan Medical University, Haikou 571199, P.R. China
| | - Xinglin Zhu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Cai Luo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yangyang Zhao
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Shanshan Pan
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Kaijia Shi
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yan Chen
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Jianmin Qiu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Zhihua Shen
- Department of Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, P.R. China.
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| | - Wei Jie
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| |
Collapse
|
6
|
Zhou QF, Yang F, Dai Y, Chen S, Zhang FR, Lu L, Lu QY. Tyrosine to threonine ratio was related to heart failure with reduced or mildly reduced ejection fraction. ESC Heart Fail 2024; 11:1567-1579. [PMID: 38361178 PMCID: PMC11098665 DOI: 10.1002/ehf2.14700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/27/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
AIMS We aim to explore the associations between serum tyrosine (Tyr) to threonine (Thr) ratio and chronic heart failure (HF) with reduced or mildly reduced ejection fraction (EF) (HFrEF or HFmrEF). METHODS AND RESULTS The study recruited 418 subjects (77.5% males, mean age 65.2 ± 12.5 years), including 318 HF subjects (HFrEF or HFmrEF) and 100 cardiovascular subjects without acute or chronic HF [including heart failure with preserved ejection fraction (HFpEF)] as controls. Serum levels of 21 kinds of amino acids (AAs) were measured by mass spectrometry. Logistic regression analysis was conducted to measuring the association between the AAs levels and the presence of HF. Event-free survival was determined by Kaplan-Meier curves and differences in survival were assessed using log-rank tests. Cox regression analysis was used to assess the prognostic value of AAs in HF. Receiver-operating characteristic (ROC) curve was performed to further confirm regression analysis. Along with the control, HFmrEF, and HFrEF subjects, serum tyrosine (Tyr) gradually increased (64.43 ± 15.28 μmol/L vs. 71.79 ± 18.74 μmol/L vs. 77.32 ± 25.90 μmol/L, P < 0.001) while serum threonine (Thr) decreased (165.21 ± 40.09 μmol/L vs. 144.93 ± 44.56 μmol/L vs. 135.25 ± 41.25 μmol/L, P < 0.001). Tyr/Thr ratio was the independent risk factor for the presence of HF in all subjects [odds ratio (OR), 3.510; 95% confidence interval (CI): 2.445-5.040; P < 0.001]. After following up for a mean year (11.10 ± 2.80 months) in 269 HF subjects (75.1% males, mean age 65.2 ± 12.8 years), the higher Tyr/Thr ratio was associated with a higher risk of HF endpoint events in HF subjects [hazard ratio (HR), 2.901; 95% CI: 1.228-6.851; P = 0.015]. By comparing the area under the receiver-operating characteristic curve (AUC), Tyr/Thr ratio was superior to Fischer's ratio (FR) in predicting HF occurrence (0.767:0.573, P < 0.001) or cardiovascular (CV) death (0.715:0.550, P = 0.047). CONCLUSIONS Circulating elevated Tyr/Thr ratio confer an increased risk for the presence of HF and poor prognosis. Tyr/Thr index outweighs FR index in predicting HF occurrence or CV death.
Collapse
Affiliation(s)
- Qing Fen Zhou
- Department of Cardiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fan Yang
- Department of Clinical Laboratory, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yang Dai
- Institution of Cardiovascular DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuai Chen
- Institution of Cardiovascular DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Feng Ru Zhang
- Department of Cardiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin Lu
- Department of Cardiology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institution of Cardiovascular DiseaseShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qiu Ya Lu
- Department of Clinical Laboratory, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Wróbel-Nowicka K, Wojciechowska C, Jacheć W, Zalewska M, Romuk E. The Role of Oxidative Stress and Inflammatory Parameters in Heart Failure. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:760. [PMID: 38792942 PMCID: PMC11123446 DOI: 10.3390/medicina60050760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024]
Abstract
Heart failure (HF) remains a major medical and social problem. The NT-pro-brain natriuretic peptide (NT-proBNP) and its active form, brain-type natriuretic peptide (BNP), in a simple blood test are the gold-standard biomarkers for HF diagnosis. However, even good biomarkers such as natriuretic peptides fail to predict all the risks associated with HF due to the diversity of the mechanisms involved. The pathophysiology of HF is determined by numerous factors, including oxidative stress, inflammation, neuroendocrine activation, pathological angiogenesis, changes in apoptotic pathways, fibrosis and vascular remodeling. High readmission and mortality rates prompt a search for new markers for the diagnosis, prognosis and treatment of HF. Oxidative-stress-mediated inflammation plays a crucial role in the development of subsequent changes in the failing heart and provides a new insight into this complex mechanism. Oxidative stress and inflammatory biomarkers appear to be a promising diagnostic and prognostic tool in patients with HF. This systematic review provides an overview of the current knowledge about oxidative stress and inflammation parameters as markers of HF.
Collapse
Affiliation(s)
- Karolina Wróbel-Nowicka
- Medical Laboratory of Teresa Fryda, Katowice, Laboratory Branch in Specialist Hospital in Zabrze, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland;
| | - Celina Wojciechowska
- 2nd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland; (C.W.); (W.J.)
| | - Wojciech Jacheć
- 2nd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland; (C.W.); (W.J.)
| | - Marzena Zalewska
- Department of Basic Medical Sciences, Faculty of Public Health in Bytom, Medical University of Silesia, Piekarska St., 41-902 Bytom, Poland;
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19, Jordan St., 41-808 Zabrze, Poland
| |
Collapse
|
8
|
Mocan D, Lala RI, Puschita M, Pilat L, Darabantiu DA, Pop-Moldovan A. The Congestion "Pandemic" in Acute Heart Failure Patients. Biomedicines 2024; 12:951. [PMID: 38790913 PMCID: PMC11117769 DOI: 10.3390/biomedicines12050951] [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: 03/01/2024] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Congestion not only represents a cardinal sign of heart failure (HF) but is also now recognized as the primary cause of hospital admissions, rehospitalization, and mortality among patients with acute heart failure (AHF). Congestion can manifest through various HF phenotypes in acute settings: volume overload, volume redistribution, or both. Recognizing the congestion phenotype is paramount, as it implies different therapeutic strategies for decongestion. Among patients with AHF, achieving complete decongestion is challenging, as more than half still experience residual congestion at discharge. Residual congestion is one of the strongest predictors of future cardiovascular events and poor outcomes. Through this review, we try to provide a better understanding of the congestion phenomenon among patients with AHF by highlighting insights into the pathophysiological mechanisms behind congestion and new diagnostic and management tools to achieve and maintain efficient decongestion.
Collapse
Affiliation(s)
- Daniela Mocan
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.)
| | - Radu Ioan Lala
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.)
- Cardiology Department, Arad County Clinical Emergency Hospital, 310037 Arad, Romania
| | - Maria Puschita
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.)
| | - Luminita Pilat
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.)
| | | | - Adina Pop-Moldovan
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.)
- Cardiology Department, Arad County Clinical Emergency Hospital, 310037 Arad, Romania
| |
Collapse
|
9
|
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
|
10
|
Wang L, Dai W, Zhu R, Long T, Zhang Z, Song Z, Mu S, Wang S, Wang H, Lei J, Zhang J, Xia W, Li G, Gao W, Zou H, Li Y, Zhan L. Testosterone and soluble ST2 as mortality predictive biomarkers in male patients with sepsis-induced cardiomyopathy. Front Med (Lausanne) 2024; 10:1278879. [PMID: 38259843 PMCID: PMC10801257 DOI: 10.3389/fmed.2023.1278879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Sepsis-induced cardiomyopathy (SIC) is characterized by high mortality and poor outcomes. This study aimed to explore the relationship between testosterone and soluble ST2 (sST2) and all-cause mortality in patients with SIC. Clinical data from SIC patients at Renmin Hospital of Wuhan University from January 2021 and March 2023 were reviewed. Serum testosterone and sST2 were measured at admission. Kaplan-Meier analysis and receiver operative characteristic curve (ROC) were used to estimate the predictive values of testosterone and sST2 on 28 days and 90 days mortality of SIC. A total of 327 male subjects with SIC were enrolled in this study. During the 28 days and 90 days follow-up, 87 (26.6%) and 103 deaths (31.5%) occurred, respectively. Kaplan-Meier analysis showed significantly higher 28 days and 90 days survival in patients with higher testosterone and decreased sST2 levels (p < 0.001). Testosterone, sST2, and N-terminal pro-B-type natriuretic peptide (NT-proBNP) were significantly associated with 28 days and 90 days mortality (p < 0.05). Partial correlation analysis showed strong positive correlation between testosterone and left ventricular ejection fraction (LVEF) (p < 0.001), and negative correlation between testosterone and sST2 (p < 0.001), high-sensitivity troponin I (hs-TnI) levels (p < 0.001) and smoke history (p < 0.01). The concentrations of sST2 were positively related with E/e' ratio (p < 0.001), and negatively correlated with TAPSE (p < 0.001). The combination of testosterone and sST2 enhanced the prediction of both 28 days [area under the ROC curve (AUC), 0.805] and 90 days mortality (AUC, 0.833). Early serum testosterone and sST2 levels could predict mortality of SIC independently and jointly. Further research is needed to determine the utility of biochemical markers in identifying high-risk patients with SIC.
Collapse
Affiliation(s)
- Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wen Dai
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruiyao Zhu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Infection Prevention and Control, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tingting Long
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sucheng Mu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shasha Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huijuan Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiaxi Lei
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Zhang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenfang Xia
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guang Li
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Li
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Shaik SP, Karan HH, Singh A, Attuluri SK, Khan AAN, Zahid F, Patil D. HFpEF: New biomarkers and their diagnostic and prognostic value. Curr Probl Cardiol 2024; 49:102155. [PMID: 37866418 DOI: 10.1016/j.cpcardiol.2023.102155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Heart failure characterized by preserved ejection fraction (HFpEF) poses a substantial challenge to healthcare systems worldwide and the diagnostic algorithms used currently mirror those utilized for reduced Ejection Fraction (HFrEF). This literature review aims to explore the diagnostic and prognostic credibility of numerous emerging biomarkers associated with HFpEF. We conducted a thorough analysis of the available medical literature and selected the biomarkers which yielded the maximum amount of published information. After reviewing the current literature we conclude that there are no biomarkers at present which are superior to natriuretic peptides in terms of diagnosis and prognosis of HFpEF. However biomarkers like Suppression of tumorigenicity2, Galectin3 and microRNAs are promising and can be researched further for future use. Although newer individual biomarkers may not be useful in diagnosing and prognosis of HFpEF, we believe that a specific biomarker profile may be identified in each phenotype,which can be used in future.
Collapse
Affiliation(s)
- Shahanaz Parveen Shaik
- Junior Resident, Internal Medicine, DR. Y.S.R University of Health Sciences, Andhra Pradesh, India.
| | - Hasnain Hyder Karan
- Resident, Internal Medicine, San Joaquin General Hospital,French Camp, CA, United States
| | - Arkaja Singh
- Junior Resident, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Sai Kiran Attuluri
- Junior Resident, Internal Medicine, DR. Y.S.R University of Health Sciences, Andhra Pradesh, India
| | - Afnan Akram Nawaz Khan
- Junior Resident, Internal Medicine, Vydehi Institute of Medical Sciences, Bangalore, India
| | - Fazila Zahid
- Resident, Internal Medicine, OSF St Francis Hospital, University of Illinois College of Medicine; IL; USA
| | - Dhrumil Patil
- Postdoctoral Research fellow, Cardiology department, Beth Israel Deaconess Medical Center, Harvard University, USA
| |
Collapse
|
12
|
Ciccarelli M, Pires IF, Bauersachs J, Bertrand L, Beauloye C, Dawson D, Hamdani N, Hilfiker-Kleiner D, van Laake LW, Lezoualc'h F, Linke WA, Lunde IG, Rainer PP, Rispoli A, Visco V, Carrizzo A, Ferro MD, Stolfo D, van der Velden J, Zacchigna S, Heymans S, Thum T, Tocchetti CG. Acute heart failure: mechanisms and pre-clinical models-a Scientific Statement of the ESC Working Group on Myocardial Function. Cardiovasc Res 2023; 119:2390-2404. [PMID: 37967390 DOI: 10.1093/cvr/cvad088] [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: 11/12/2022] [Revised: 02/16/2023] [Accepted: 03/06/2023] [Indexed: 11/17/2023] Open
Abstract
While chronic heart failure (CHF) treatment has considerably improved patient prognosis and survival, the therapeutic management of acute heart failure (AHF) has remained virtually unchanged in the last decades. This is partly due to the scarcity of pre-clinical models for the pathophysiological assessment and, consequently, the limited knowledge of molecular mechanisms involved in the different AHF phenotypes. This scientific statement outlines the different trajectories from acute to CHF originating from the interaction between aetiology, genetic and environmental factors, and comorbidities. Furthermore, we discuss the potential molecular targets capable of unveiling new therapeutic perspectives to improve the outcome of the acute phase and counteracting the evolution towards CHF.
Collapse
Affiliation(s)
- Michele Ciccarelli
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081 Baronissi, Italy
| | - Inês Falcão Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christophe Beauloye
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Dana Dawson
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St.Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Linda W van Laake
- Division Heart and Lungs, Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Frank Lezoualc'h
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Robert-Koch-Str. 27B, Münster 48149, Germany
| | - Ida G Lunde
- Division of Diagnostics and Technology (DDT), Akershus University Hospital, and KG Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- BioTechMed Graz - University of Graz, 8036 Graz, Austria
| | - Antonella Rispoli
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081 Baronissi, Italy
| | - Valeria Visco
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081 Baronissi, Italy
| | - Albino Carrizzo
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Via Salvador Allende, 84081 Baronissi, Italy
- Laboratory of Vascular Physiopathology-I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy
| | - Matteo Dal Ferro
- Cardiothoracovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
- Laboratory of Cardiovascular Biology, The International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Davide Stolfo
- Cardiothoracovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, Netherlands
| | - Serena Zacchigna
- Laboratory of Cardiovascular Biology, The International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental medicine, Hannover, Germany
| | - Carlo Gabriele Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
13
|
Durak-Nalbantic A, Begic E, Begic A, Dzubur A, Lepara O, Baljic R, Hamzic-Mehmedbasic A, Rebic D, Hodzic E, Halimic M, Badnjevic A. Biomarkers and their combination in a prediction of decompensation after an index hospitalization for acute heart failure. J Family Med Prim Care 2023; 12:1158-1164. [PMID: 37636186 PMCID: PMC10451570 DOI: 10.4103/jfmpc.jfmpc_1456_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/29/2022] [Accepted: 02/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Heart failure (HF) still remains as one of the most common causes of hospital admission with a high mortality rate. Aim To investigate the possible prognostic role of brain natriuretic peptide (BNP), high-sensitivity (hs) cardiac troponin (cTn) I, cystatin C, and cancer antigen 125 (CA125) in the prediction of decompensation after an index hospitalization and to investigate their possible additive prognostic value. Patients and Methods Two hundred twenty-two patients hospitalized with acute HF were monitored and followed for 18 months. Results BNP at discharge has the highest sensitivity and specificity in the prediction of decompensation. For a cutoff value of 423.3 pg/ml, sensitivity was 64.3% and specificity was 64.5%, with a positive predictive value of 71.6% and an area under the curve (AUC) of 0.69 (P < 0.001). The hazard risk (HR) for decompensation when the discharge BNP was above the cutoff value was 2.18. Cystatin C, at a cutoff value of 1.46 mg/L, had a sensitivity of 57% and specificity of 57.8%, with a positive predictive value of 65.8% and an AUC of 0.59 (P = 0.028). CA125, in the prediction of decompensation in patients with acute heart failure (AHF) and at a cutoff value of 80.5 IU/L, had a sensitivity of 60.5% and specificity of 53.3%, with a positive predictive value of 64.5% and an AUC of 0.59 (P = 0.022). The time till onset of decompensation was significantly shorter in patients with four versus three elevated biomarkers (P = 0.047), with five versus three elevated biomarkers (P = 0.026), and in patients with four versus two elevated biomarkers (P = 0.026). The HR for decompensation in patients with five positive biomarkers was 3.7 (P = 0.001) and in patients with four positive biomarkers was 2.5 (P = 0.014), compared to patients who had fewer positive biomarkers. Conclusion BNP, cystatin C, and CA125 are predictors of decompensation, and their combined usage leads to better prediction of new decompensation.
Collapse
Affiliation(s)
- Azra Durak-Nalbantic
- Clinic for Heart and Vessel Disease and Rheumatism, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Edin Begic
- Medical School, Sarajevo School of Science and Tecnology, Hrasnička Cesta 3a, Sarajevo, Bosnia and Herzegovina
| | - Alden Begic
- Clinic for Heart and Vessel Disease and Rheumatism, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Alen Dzubur
- Clinic for Heart and Vessel Disease and Rheumatism, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Orhan Lepara
- Medical Faculty, University of Sarajevo, Cekalusa 90, Sarajevo, Bosnia and Herzegovina
| | - Rusmir Baljic
- Clinic for Infective Disease, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Aida Hamzic-Mehmedbasic
- Medical School, Sarajevo School of Science and Tecnology, Hrasnička Cesta 3a, Sarajevo, Bosnia and Herzegovina
| | - Damir Rebic
- Clinic for Nephrology, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Enisa Hodzic
- Clinic for Heart and Vessel Disease and Rheumatism, Clinical Center University of Sarajevo, Bolnicka 25, Sarajevo, Bosnia and Herzegovina
| | - Mirza Halimic
- Pediatric Clinic, Clinical Center University of Sarajevo, Patriotske Lige, 87, Sarajevo, Bosnia and Herzegovina
| | - Almir Badnjevic
- International Burch University, Faculty of Engineering and Natural Sciences, Genetics and Bioengineering Department, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
14
|
Berezin AE, Berezin AA. Biomarkers in Heart Failure: From Research to Clinical Practice. Ann Lab Med 2023; 43:225-236. [PMID: 36544334 DOI: 10.3343/alm.2023.43.3.225] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/19/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022] Open
Abstract
The aim of this narrative review is to summarize contemporary evidence on the use of circulating cardiac biomarkers of heart failure (HF) and to identify a promising biomarker model for clinical use in personalized point-of-care HF management. We discuss the reported biomarkers of HF classified into clusters, including myocardial stretch and biomechanical stress; cardiac myocyte injury; systemic, adipocyte tissue, and microvascular inflammation; cardiac fibrosis and matrix remodeling; neurohumoral activation and oxidative stress; impaired endothelial function and integrity; and renal and skeletal muscle dysfunction. We focus on the benefits and drawbacks of biomarker-guided assistance in daily clinical management of patients with HF. In addition, we provide clear information on the role of alternative biomarkers and future directions with the aim of improving the predictive ability and reproducibility of multiple biomarker models and advancing genomic, transcriptomic, proteomic, and metabolomic evaluations.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, Zaporozhye Medical Academy of Postgraduate Education, Zaporozhye, Ukraine
| | - Alexander A Berezin
- Internal Medicine Department, Zaporozhye Medical Academy of Postgraduate Education, Zaporozhye, Ukraine
| |
Collapse
|
15
|
The soluble suppression of tumorigenicity 2 as a biomarker of early cardiac remodeling in bradycardia patients receiving permanent pacemaker therapy. Future Sci OA 2023; 9:FSO831. [PMID: 37006226 PMCID: PMC10051037 DOI: 10.2144/fsoa-2023-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Aim: This study aims to evaluate: the difference of soluble suppression of tumorigenicity 2 (sST2) level, a biomarker for cardiac remodeling and echocardiography parameters value prior to and 1 month after implantation; and the association between pacemaker parameters and pacemaker mode along with delta sST2 levels. Materials & methods: This prospective cohort study enrolled all symptomatic bradycardia patients aged >18 years with preserved ejection fraction who underwent permanent pacemaker (PPM) implantation. Results: A total of 49 patients were included in this study. The sST2 level (ng/ml) were significantly different between prior and 1 month following PPM implantation (23.4 ± 28.4 vs 39.9 ± 63.7; p = 0.001). Conclusion: The early cardiac remodeling has occurred within 1 month after PPM implantation as indicated by increasing delta sST2 level.
Collapse
|
16
|
Chhor M, Law W, Pavlovic M, Aksentijevic D, McGrath K, McClements L. Diagnostic and prognostic biomarkers reflective of cardiac remodelling in diabetes mellitus: A scoping review. Diabet Med 2023; 40:e15064. [PMID: 36782075 DOI: 10.1111/dme.15064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 01/19/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
AIMS The aim of this scoping review is to evaluate the current biomarkers used in the assessment of adverse cardiac remodelling in people with diabetes mellitus (DM) and in the diagnosis and prognosis of subsequent cardiovascular disease. We aim to discuss the biomarkers' pathophysiological roles as a reflection of the cardiac remodelling mechanisms in the presence of DM. METHODS We performed the literature search to include studies from 2003 to 2021 using the following databases: MEDLINE, Scopus, Web of Science, PubMed, and Cochrane library. Articles that met our inclusion criteria were screened and appraised before being included in this review. The PRISMA guidelines for Scoping Reviews were followed. RESULTS Our literature search identified a total of 43 eligible articles, which were included in this scoping review. We identified 15 different biomarkers, each described by at least two studies, that were used to determine signs of cardiac remodelling in cardiovascular disease (CVD) and people with DM. NT-proBNP was identified as the most frequently employed biomarker in this context; however, we also identified emerging biomarkers including hs-CRP, hs-cTnT, and Galectin-3. CONCLUSION There is a complex relationship between DM and cardiovascular health, where more research is needed. Current biomarkers reflective of adverse cardiac remodelling in DM are often used to diagnose other CVDs, such as NT-proBNP for heart failure. Hence there is a need for identification of specific biomarkers that can detect early signs of cardiac remodelling in the presence of DM. Further research into these biomarkers and mechanisms can deepen our understanding of their role in DM-associated CVD and lead to better preventative therapies.
Collapse
Affiliation(s)
- Michael Chhor
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Sydney, Australia
| | - William Law
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Sydney, Australia
| | - Milan Pavlovic
- Department of Internal Medicine - Cardiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Sydney, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Sydney, Australia
| |
Collapse
|
17
|
Jia Y, Li D, Yu J, Jiang W, Liu Y, Li F, Li W, Zeng R, Liao X, Wan Z. Prognostic value of interleukin-33, sST2, myeloperoxidase, and matrix metalloproteinase-9 in acute aortic dissection. Front Cardiovasc Med 2023; 9:1084321. [PMID: 36684579 PMCID: PMC9853981 DOI: 10.3389/fcvm.2022.1084321] [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: 10/30/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Background and purpose Acute aortic dissection (AAD) is a life-threatening cardiovascular emergency. Both neutrophil granzyme and interleukin (IL)-33/ST2 systems have proven to be effective diagnostic markers for AAD. This study aimed to investigate the relationship between plasma IL-33, soluble suppression of tumorigenesis-2 (sST2), myeloperoxidase (MPO), and matrix metalloproteinase (MMP)-9 levels at admission and all-cause mortality in patients with AAD. Methods A total of 155 patients with AAD were enrolled from the Prospective Evaluation of Acute Chest Pain (PEACP) study. Plasma concentrations of IL-33, sST2, and MMP-9 were measured using an enzyme-linked immunosorbent assay, and MPO was detected using a chemiluminescence immunoassay. Aortic anatomical parameters were measured using CT radiography. The primary endpoint was all-cause mortality rate. Results The median age of the patients was 55 years, and 96 (61.9%) were diagnosed with type A-AAD. After adjusting for confounding factors, the highest tertiles of IL-33, sST2, MPO, and MMP-9 had hazard risks of 0.870 (95% CI: 0.412-1.836, P = 0.714), 3.769 (95% CI: 1.504-9.446, P = 0.005), 4.689 (95% CI: 1.985-11.076, P < 0.001), and 4.748 (95% CI: 1.763-12.784, P = 0.002), respectively, compared to the lowest tertile. Pearson's correlation analysis revealed a significant correlation between these markers (P < 0.001). Moreover, sST2, MPO, and MMP-9 levels had a significant positive correlation with aortic diameter and pseudolumen area (P < 0.001). Conclusion The biomarkers sST2, MPO, and MMP-9 were independently associated with mortality in patients with AAD. The significant correlation between these biomarkers suggests a pathogenic role for the IL-33/ST2/neutrophil granzyme system in patients with AAD.
Collapse
Affiliation(s)
- Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Jing Yu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Fanghui Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wentao Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Zeng
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhi Wan
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Zhi Wan,
| |
Collapse
|
18
|
Riley ED, Kazi DS, Coffin PO, Vittinghoff E, Wade AN, Bulfone TC, Lynch KL, Atai Z, Wu AH. Impact of multiple substance use on circulating ST2, a biomarker of adverse cardiac remodelling, in women. Biomarkers 2022; 27:802-808. [PMID: 36168954 PMCID: PMC9744090 DOI: 10.1080/1354750x.2022.2129451] [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: 04/29/2022] [Accepted: 09/22/2022] [Indexed: 01/28/2023]
Abstract
CONTEXT Cardiovascular disease (CVD) and heart failure (HF) are major causes of mortality in low-income populations and differ by sex. Risk assessment that incorporates cardiac biomarkers is common. However, research evaluating the utility of biomarkers rarely includes controlled substances, which may influence biomarker levels and thus influence CVD risk assessment. MATERIALS AND METHODS We identified the effects of multiple substances on soluble "suppression of tumorigenicity 2" (sST2), a biomarker of adverse cardiac remodelling, in 245 low-income women. Adjusting for CVD risk factors, we examined associations between substance use and sST2 over six monthly visits. RESULTS Median age was 53 years and 74% of participants were ethnic minority women. An sST2 level > 35 ng/mL (suggesting cardiac remodelling) during ≥1 study visit was observed in 44% of participants. In adjusted analysis, higher sST2 levels were significantly and positively associated with the presence of cocaine (Adjusted Linear Effect [ALE]:1.10; 95% CI:1.03-1.19), alcohol (ALE:1.10; 95% CI:1.04-1.17), heroin (ALE:1.25; 95% CI:1.10-1.43), and the interaction between heroin and fentanyl use. CONCLUSION Results suggest that the use of multiple substances influences the level of sST2, a biomarker often used to evaluate cardiovascular risk. Incorporating substance use alongside cardiac biomarkers may improve CVD risk assessment in vulnerable women.
Collapse
Affiliation(s)
- Elise D. Riley
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, California, USA
| | - Dhruv S. Kazi
- Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA and Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Phillip O. Coffin
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, California, USA
- San Francisco Department of Public Health, San Francisco, California, USA
| | - Eric Vittinghoff
- University of California, San Francisco, School of Medicine, Department of Epidemiology and Biostatistics, San Francisco, California, USA
| | - Amanda N. Wade
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, California, USA
| | - Tommaso C. Bulfone
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, California, USA
| | - Kara L. Lynch
- University of California, San Francisco, School of Medicine, Department of Laboratory Medicine, San Francisco, California, USA
| | - Zahra Atai
- University of California, San Francisco, School of Medicine, Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, San Francisco, California, USA
| | - Alan H.B. Wu
- University of California, San Francisco, School of Medicine, Department of Laboratory Medicine, San Francisco, California, USA
| |
Collapse
|
19
|
Shi Y, Liu J, Liu C, Shuang X, Yang C, Qiao W, Dong G. Diagnostic and prognostic value of serum soluble suppression of tumorigenicity-2 in heart failure with preserved ejection fraction: A systematic review and meta-analysis. Front Cardiovasc Med 2022; 9:937291. [PMID: 36204571 PMCID: PMC9530661 DOI: 10.3389/fcvm.2022.937291] [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: 05/06/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background Heart failure (HF) with preserved ejection fraction (HFpEF) is a growing public health burden, with mortality and rehospitalization rates comparable to HF with reduced ejection fraction (HFrEF). The evidence for the clinical usefulness of soluble suppression of tumorigenicity 2 (sST2) in HFpEF is contradictory. Therefore, we conducted the following systematic review and meta-analysis to assess the diagnostic and prognostic value of serum sST2 in HFpEF. Methods PubMed and Scopus were searched exhaustively from their inception until March 15, 2022. In diagnostic analysis, we compared the diagnostic value of serum sST2 in HFpEF to NT pro-BNP. We separately pooled the unadjusted and multivariate-adjusted hazard ratios (HRs) and the corresponding 95% confidence intervals (CIs) in prognostic analysis. Results A total of 16 publications from 2008 to 2021 were examined. The results of this analysis were as follow: Firstly, compared with NT pro-BNP, sST2 obtains poor diagnostic performance in independently identifying HFpEF from healthy controls, hypertensive patients, and HFrEF patient. Nevertheless, it may provide incremental value to other biomarkers for diagnosing HFpEF and deserves further investigation. Secondly, log sST2 was independently associated with adverse endpoints on multivariable analysis after adjusting for variables such as age, sex, race, and NYHA class. Per log unit rise in sST2, there was a 2.76-fold increased risk of all-cause death [HR:2.76; 95% CI (1.24, 6.16); p = 0.516, I2 = 0%; P = 0.013] and a 6.52-fold increased risk in the composite endpoint of all-cause death and HF hospitalization [HR:6.52; 95% CI (2.34, 18.19); p = 0.985, I2 = 0%; P = 0.000]. Finally, the optimal threshold levels of serum sST2 need further determined. Conclusions Higher sST2 was strongly linked to an increased risk of adverse outcomes in HFpEE. Especially, log sST2 independently predicted all-cause death and the composite endpoint of all-cause death and HF hospitalization. However, prospective and multicenter studies with large-sample and extended follow-up periods are required to validate our results due to limitations in our research.
Collapse
Affiliation(s)
- Yujiao Shi
- Department of Post-graduate Institute, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jiangang Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Chunqiu Liu
- Department of Post-graduate Institute, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Xiong Shuang
- Department of Post-graduate Institute, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Chenguang Yang
- Department of Post-graduate Institute, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenbo Qiao
- Department of Post-graduate Institute, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Guoju Dong
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- *Correspondence: Guoju Dong
| |
Collapse
|
20
|
Ponikowska B, Iwanek G, Zdanowicz A, Urban S, Zymliński R, Ponikowski P, Biegus J. Biomarkers of Myocardial Injury and Remodeling in Heart Failure. J Pers Med 2022; 12:799. [PMID: 35629221 PMCID: PMC9144334 DOI: 10.3390/jpm12050799] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/08/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
With its complicated pathophysiology, high incidence and prevalence, heart failure remains a major public concern. In hopes of improving diagnosis, treatment and prognosis, the utility of many different biomarkers is researched vigorously around the world. In this review, biomarkers of myocardial remodeling and fibrosis (galectin-3, soluble isoform of suppression of tumorigenicity 2, matrix metalloproteinases, osteopontin, interleukin-6, syndecan-4, myostatin, procollagen type I C-terminal propeptide, procollagen type III N-terminal propeptide, vascular endothelial growth factor, nitric oxidase synthetase and asymmetric dimethylarginine), myocyte injury (heart-type fatty acid-binding protein, glutathione S-transferase P1 and heat shock protein 60), as well as iron metabolism (ferritin, transferrin saturation, soluble transferrin receptor and hepcidin), are considered in terms of possible clinical applicability and significance. Our short review consists of a summary of the aforementioned cardiovascular biomarkers' clinical relevance and perspectives.
Collapse
Affiliation(s)
- Barbara Ponikowska
- Student Scientific Organization, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Gracjan Iwanek
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| | - Agata Zdanowicz
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| | - Szymon Urban
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| | - Robert Zymliński
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (A.Z.); (S.U.); (R.Z.); (P.P.); (J.B.)
| |
Collapse
|
21
|
Soluble ST2 as a Potential Biomarker for Risk Assessment of Pulmonary Hypertension in Patients Undergoing TAVR? Life (Basel) 2022; 12:life12030389. [PMID: 35330140 PMCID: PMC8954652 DOI: 10.3390/life12030389] [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: 02/11/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Severe aortic valve stenosis (AS) is associated with pulmonary hypertension (PH) and has been shown to limit patient survival. Soluble suppression of tumorigenicity-2 (sST2) is a cardiovascular biomarker that has proven to be an important prognostic marker for survival in patients undergoing transcatheter aortic valve replacement (TAVR). The aim of this study was to assess the importance of the sST2 biomarker for risk stratification in patients with severe AS in presence or absence of PH. Methods: In 260 patients with severe AS undergoing TAVR procedure, sST2 serum level concentrations were analyzed. Right heart catheter measurements were performed in 152 patients, with no PH detection in 43 patients and with PH detection in 109 patients. Correlation analyses according to Spearman, AUROC analyses and Kaplan–Meier curves were calculated. Results: Patients with severe AS and PH showed significantly higher serum sST2 concentrations (p = 0.006). The sST2 cut-off value for non-PH patients regarding 1-year survival yielded 5521.15 pg/mL, whereas the cut-off value of PH patients was at a considerably higher level of 10,268.78 pg/mL. A cut-off value of 6990.12 pg/mL was related with a significant probability of PH presence. Survival curves showed that patients with severe AS and PH not only had higher 1-year mortality, but also that increased levels of sST2 plasma concentration were associated with earlier death. Conclusion: sST2 definitely has the potential to provide information about the presence of PH in patients with severe AS, in a noninvasive way.
Collapse
|
22
|
Correlation between Serum LP-PLA2 and sST2 Levels and the Condition of Patients with Acute Heart Failure and Their Prognostic Value. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8267776. [PMID: 34707676 PMCID: PMC8545552 DOI: 10.1155/2021/8267776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/27/2021] [Indexed: 12/01/2022]
Abstract
Acute heart failure (AHF) occurs mostly in the elderly, which is a syndrome that occurs in the later stages of the development of cardiovascular disease. Due to the sharp decline in the patient's heart function, the patient has a high mortality rate and a poor prognosis, which seriously threaten the life safety of the elderly. Therefore, early diagnosis of AHF and timely treatment are extremely important. Lipoprotein-associated phospholipase A2 (LP-PLA2) is a newly discovered cardiovascular-specific inflammatory marker, which is closely related to the formation and progression of atherosclerotic plaque and the occurrence and development of coronary heart disease. Soluble growth stimulation expression gene 2 (sST2) protein is a protein produced under the induction of mechanical stress in cardiomyocytes. It can act as a decoy receptor to mediate the interleukin-33 (IL-33)/ST2 signaling pathway to bind to IL-33, thereby reducing the myocardial protective effect and leading to myocardial remodeling. The purpose of this study was to explore the serum LP-PLA2 and sST2 levels in AHF patients and to analyze their correlation with the disease and their prognostic value. The results showed that the levels of serum LP-PLA2 and sST2 were increased in AHF patients, and the levels of serum LP-PLA2 and sST2 in patients with adverse prognostic events were higher than those in patients without adverse prognostic events. The levels of serum LP-PLA2 and sST2 are closely related to the degree of patients' illness, among which the combined prediction of AHF patients with LP-PLA2 and sST2 has the highest value, which is worthy of promotion.
Collapse
|
23
|
Levels of Serum sST2, MMP-3, and Gal-3 in Patients with Essential Hypertension and Their Correlation with Left Ventricular Hypertrophy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7262776. [PMID: 34675989 PMCID: PMC8526212 DOI: 10.1155/2021/7262776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 12/17/2022]
Abstract
Essential hypertension (EH) is a clinically frequent cardiovascular disease, with insidious onset, causing increased pressure load and neuroregulation disorders in patients. Long-term EH can cause left ventricular hypertrophy (LVH), which can lead to arrhythmia and even death. The soluble suppression of tumorigenicity 2 (sST2), matrix metalloproteinase-3 (MMP-3), and galectin-3 (Gal-3) in serum plays an important role in the occurrence, development, and prognosis of cardiovascular diseases. In our study, we divided EH patients into 3 levels and groups with or without LVH, according to their condition. The levels of sST2, MMP-3, and Gal-3 in the serum were measured in different groups of patients. Our results showed that the levels of sST2, MMP-3, and Gal-3 in the serum increased progressively with the level in different EH groups. The levels of sST2, MMP-3, and Gal-3 in the serum of the LVH group were higher than those of the NLVH group, and it is positively correlated with LVH-related indexes. The risk of developing and progressing to LVH in patients with EH can be determined by the method of measuring three indicators.
Collapse
|
24
|
A Novel Paradigm Based on ST2 and Its Contribution towards a Multimarker Approach in the Diagnosis and Prognosis of Heart Failure: A Prospective Study during the Pandemic Storm. Life (Basel) 2021; 11:life11101080. [PMID: 34685450 PMCID: PMC8539225 DOI: 10.3390/life11101080] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/03/2021] [Accepted: 10/09/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Acute heart failure (HF) represents an increasingly common and challenging presentation in the emergency room, also inducing a great socio-economic burden. Extensive research was conducted toward finding an ideal biomarker of acute HF, both in terms of sensitivity and specificity, but today practicians’ interest has shifted towards a more realistic multimarker approach. Natriuretic peptides (NPs) currently represent the gold standard for diagnosing HF in routine clinical practice, but novel molecules, such as sST2, emerge as potentially useful biomarkers, providing additional diagnostic and prognostic value. Methods: We conducted a prospective, single-center study that included 120 patients with acute HF and 53 controls with chronic HF. Of these, 13 patients (eight with acute HF, five from the control group) associated the coronavirus-19 disease (COVID-19). The diagnosis of HF was confirmed by a complete clinical, biological and echocardiographic approach. Results: The serum levels of all studied biomarkers (sST2, NT-proBNP, cardiac troponin) were significantly higher in the group with acute HF. By area under the curve (AUC) analysis, we noticed that NT-proBNP (AUC: 0.976) still had the best diagnostic performance, closely followed by sST2 (AUC: 0.889). However, sST2 was a significantly better predictor of fatal events, showing positive correlations for both in-hospital and at 1-month mortality rates. Moreover, sST2 was also associated with other markers of poor prognosis, such as the use of inotropes or high lactate levels, but not with left ventricle ejection fraction, age, body mass index or mean arterial pressure. sST2 levels were higher in patients with a positive history of COVID-19 as compared with non-COVID-19 patients, but the differences were statistically significant only within the control group. Bivariate regression showed a positive and linear relationship between NT-proBNP and sST2 (r(120) = 0.20, p < 0.002). Conclusions: we consider that sST2 has certain qualities worth integrating in a future multimarker test kit alongside traditional biomarkers, as it provides similar diagnostic value as NT-proBNP, but is emerging as a more valuable prognostic factor, with a better predictive value of fatal events in patients with acute HF.
Collapse
|
25
|
Jin Y, Wei S, Yao L. Diagnostic performance of miR-214, BNP, NT-proBNP and soluble ST2 in acute heart failure. Int J Clin Pract 2021; 75:e14643. [PMID: 34310833 DOI: 10.1111/ijcp.14643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/23/2021] [Indexed: 11/28/2022] Open
Abstract
PURPOSE This study aimed to investigate the potential diagnostic value of miR-214, B-type natriuretic peptide (BNP), N terminal-pro BNP (NT-proBNP) and soluble ST2 (sST2) in acute heart failure (AHF). METHOD This study included 176 patients as the AHF group and 60 healthy subjects as the control group from February 2018 to February 2020. Patients in the AHF group were classified according to the New York Heart Association (NYHA) functional classification, including 60 level II patients, 59 level III patients and 57 level IV patients. The expression level of miR-214, BNP, NT-proBNP and sST2 of both groups were recorded and analysed. RESULTS The morbidity of cardiovascular diseases was significantly higher in the AHF group than in the control group (P < .05). The expression level of miR-214, BNP, NT-proBNP and sST2 in the AHF group were all significantly higher than in the control group (P < .05). Besides, the expression level of all the molecules in level IV was significantly higher than that of level III and level II, respectively (P < .001, P < .001). In addition, the expression level of all the molecules in level III was significantly higher than that of level II (P < .001). The area under the ROC curve of miR-214, BNP, NT-proBNP and sST2 were 0.913, 0.836, 0.849 and 0.855, respectively, indicating good diagnostic value. CONCLUSION MiR-214, BNP, NT-proBNP and sST2 can be used as effective biomarkers for AHF, providing a new strategy for diagnosis and for judging the severity of AHF.
Collapse
Affiliation(s)
- Yinsheng Jin
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Shuang Wei
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Lingling Yao
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
26
|
Liu Y, Gao X, Xiao Q, Wang W, Zhu B. Correlation Between QTc Dispersion and Soluble Growth-stimulating Gene 2 Protein on the Early Prognosis of Acute Carbon Monoxide Poisoning Heart Disease. J Cardiovasc Pharmacol 2021; 78:572-580. [PMID: 34166304 DOI: 10.1097/fjc.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/05/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT This study aimed to explore the correlation between QTc dispersion (QTcd) and soluble growth-stimulating gene 2 protein (sST2) after heart rate correction in patients with acute carbon monoxide poisoning heart disease. Among the 150 patients, 35 cases had severe toxic heart disease. The concentrations of sST2, cardiac troponin I, and creatine kinase-MB in the severe group began to increase from admission, 24 hours, and 2 days, respectively, and their detected values were all higher than those in the nonsevere group and the normal control group. There were statistically significant differences in sST2 and QTcd between the poisoning, nonsevere, and normal control groups before the treatment. There was a statistically significant difference between the indexes of the poisoning groups at different degrees 2 and 3 days after poisoning. Receiver operating characteristic curve analysis confirmed the sensitivity and specificity of sST2 and QTcd. The correlation analysis showed that sST2 and QTcd levels were positively correlated with the incidence of severe heart disease at admission. Generally, the combined observation of sST2 and QTcd improved the prediction sensitivity and were early predictor indexes of toxic heart disease.
Collapse
Affiliation(s)
- Yongjian Liu
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | | | | | | | | |
Collapse
|
27
|
Could a Multi-Marker and Machine Learning Approach Help Stratify Patients with Heart Failure? MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57100996. [PMID: 34684033 PMCID: PMC8538712 DOI: 10.3390/medicina57100996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022]
Abstract
Half of the patients with heart failure (HF) have preserved ejection fraction (HFpEF). To date, there are no specific markers to distinguish this subgroup. The main objective of this work was to stratify HF patients using current biochemical markers coupled with clinical data. The cohort study included HFpEF (n = 24) and heart failure with reduced ejection fraction (HFrEF) (n = 34) patients as usually considered in clinical practice based on cardiac imaging (EF ≥ 50% for HFpEF; EF < 50% for HFrEF). Routine blood tests consisted of measuring biomarkers of renal and heart functions, inflammation, and iron metabolism. A multi-test approach and analysis of peripheral blood samples aimed to establish a computerized Machine Learning strategy to provide a blood signature to distinguish HFpEF and HFrEF. Based on logistic regression, demographic characteristics and clinical biomarkers showed no statistical significance to differentiate the HFpEF and HFrEF patient subgroups. Hence a multivariate factorial discriminant analysis, performed blindly using the data set, allowed us to stratify the two HF groups. Consequently, a Machine Learning (ML) strategy was developed using the same variables in a genetic algorithm approach. ML provided very encouraging explorative results when considering the small size of the samples applied. The accuracy and the sensitivity were high for both validation and test groups (69% and 100%, 64% and 75%, respectively). Sensitivity was 100% for the validation and 75% for the test group, whereas specificity was 44% and 55% for the validation and test groups because of the small number of samples. Lastly, the precision was acceptable, with 58% in the validation and 60% in the test group. Combining biochemical and clinical markers is an excellent entry to develop a computer classification tool to diagnose HFpEF. This translational approach is a springboard for improving new personalized treatment methods and identifying “high-yield” populations for clinical trials.
Collapse
|
28
|
Harmon DM, AbouEzzeddine OF, McKie PM, Scott CG, Saenger AK, Jaffe AS. Sex-specific cut-off values for soluble suppression of tumorigenicity 2 (ST2) biomarker increase its cardiovascular prognostic value in the community. Biomarkers 2021; 26:639-646. [PMID: 34269635 DOI: 10.1080/1354750x.2021.1956590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Suppression of tumorigenicity 2 (ST2) has important cardiovascular prognostic value in community patients; however, previous analyses have utilized non-sex specific cut-off values. We assessed whether sex-specific ST2 cut-off values would improve the prognostic utility of ST2 in the asymptomatic community. METHODS A total of 2042 participants underwent clinical assessment and echocardiographic evaluation. Baseline measurements of high sensitivity troponin, natriuretic peptides and ST2 were obtained in 1681 individuals. ST2, cardiac biomarkers and associated co-morbidities were evaluated by sex-specific ST2 quartile analysis. ST2 concentrations were also analysed as dichotomous variables defined as being above the sex-specific cut-off for each the outcomes of heart failure (HF), major adverse cardiac event (MACE) and mortality. RESULTS Median ST2 concentration was 29.4 ng/mL in male subjects and 24.1 ng/mL in female subjects. Higher ST2 concentrations were associated with incident HF (p<0.001; preserved ejection fraction (EF) p<0.001, reduced EF p=0.23), MACE (p=0.003) and mortality (p<0.001) across sex-specific quartiles. Event-based, hazard ratio (HR) analysis revealed sex-specific ST2 cut-offs were significantly more predictive of incident HF, MACE and mortality compared to non-sex-specific analysis even following adjustment for cardiac co-morbidities and traditional biomarkers. CONCLUSIONS These data suggest that sex-specific cut-offs, greater than non-sex specific cut-offs, significantly impact the prognostic value of the biomarker ST2 in the asymptomatic community cohort.Clinical SignificanceSuppression of tumorigenicity 2 (ST2) is a biomarker which has known associations with heart failure (HF), major adverse cardiac events (MACEs) and mortality in the general population.Recent data support the concept of sex-specific cut off values and individualized approaches based on sex to predict cardiovascular disease. Given the difference in pathobiology between the sexes, the fact that such approaches improve risk stratification is understandable. Thus, when sex-specific treatments are developed, this may similarly lead to improved outcomes.The use of sex-specific ST2 cut-off values significantly improved the prognostic value in predicting HF, MACE, and mortality in an asymptomatic community. This prognostication was particularly strong for HF with preserved ejection fraction and remained clinically significant following adjustment for cardiac co-morbidities and other traditional cardiac biomarkers (NTproBNP and hscTnI).
Collapse
Affiliation(s)
- David M Harmon
- Department of Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN, USA
| | | | - Paul M McKie
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Amy K Saenger
- Department of Laboratory Medicine and Pathology, Hennepin Healthcare/HCMC, Minneapolis, MN, USA.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Allan S Jaffe
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
29
|
Yang C, Fan Z, Wu J, Zhang J, Zhang W, Yang J, Yang J. The Diagnostic Value of Soluble ST2 in Heart Failure: A Meta-Analysis. Front Cardiovasc Med 2021; 8:685904. [PMID: 34327224 PMCID: PMC8315235 DOI: 10.3389/fcvm.2021.685904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/10/2021] [Indexed: 01/09/2023] Open
Abstract
Objective: The diagnostic performance of soluble suppression of tumorigenicity (sST2) in heart failure (HF) had been investigated in multiple studies, but the results were inconsistent. This meta-analysis evaluated the diagnostic value of sST2 in HF. Methods: Pubmed, Web of Science, Embase, and Cochrane Library databases were searched until March 2021. Cohort studies or case-control studies relevant to the diagnostic value of sST2 in HF were screened, and true positive (TP), false positive (FP), false negative (FN), and true negative (TN) data were extracted for calculating sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC). The quality of the included studies was evaluated using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS), the threshold effect was determined by calculating Spearman correlation coefficients and summary receiver operating characteristic (SROC) curve patterns, the heterogeneity was evaluated using the I2 statistic and the Galbraith radial plot, and sensitivity analysis was also performed. Deeks' test was used to assess publication bias. Results: A total of 11 studies from 10 articles were included in this meta-analysis. The Spearman correlation coefficient was 0.114, p = 0.739, and the SROC curve did not show a “shoulder-arm” shape, which suggests that there was no threshold effect, but study heterogeneity existed because of non-threshold effects. The combined sensitivity was 0.72 [95% confidence interval (CI): 0.65–0.78], specificity was 0.65 (95% CI: 0.45–0.81), PLR was 1.75 (95% CI: 1.33–2.31), NLR was 0.48 (95% CI: 0.37–0.63), DOR was 3.63 (95% CI: 2.29–5.74), and AUC was 0.75. The Deeks' test suggested no significant publication bias in the included studies (P = 0.94). Conclusion: sST has some diagnostic value in HF, but this should be further evaluated in additional studies with rigorous design and high homogeneity.
Collapse
Affiliation(s)
- Chaojun Yang
- Central Laboratory, Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University and Yichang Central People's Hospital, Yichang, China
| | - Zhixing Fan
- Central Laboratory, Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University and Yichang Central People's Hospital, Yichang, China
| | - Jinchun Wu
- Department of Cardiology, Qinghai Provincial People's Hospital, Xining, China
| | - Jing Zhang
- Central Laboratory, Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University and Yichang Central People's Hospital, Yichang, China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Yang
- Department of Cardiology, The People's Hospital of Three Gorges University and The First People' s Hospital of Yichang, Yichang, China
| | - Jun Yang
- Central Laboratory, Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University and Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
30
|
Wang QG, Cheng BCY, He YZ, Li LJ, Ling Y, Luo G, Wang L, Liang S, Zhang Y. miR-320a in serum exosomes promotes myocardial fibroblast proliferation via regulating the PIK3CA/Akt/mTOR signaling pathway in HEH2 cells. Exp Ther Med 2021; 22:873. [PMID: 34194551 PMCID: PMC8237386 DOI: 10.3892/etm.2021.10305] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) serve an important role in the pathogenesis of chronic heart failure (CHF). A number of reports have illustrated the regulatory effect of serum exosomal miRNA on myocardial fibrosis. The present study aimed to investigate the expression of miR-320a in serum exosomes, as well as the effect of miR-320a on myocardial fibroblast proliferation. Serum exosome samples from 10 patients with CHF and 5 healthy volunteers were obtained and characterized. mRNA and protein expression levels were measured via reverse transcription-quantitative PCR and western blotting, respectively. The content of soluble growth stimulation expressed gene 2 (sST2) was determined via ELISA. HEH2 cell viability and apoptosis were detected by performing MTT assays and flow cytometry, respectively. The results demonstrated that serum miR-320a expression levels and sST2 content were significantly increased in patients with CHF compared with healthy controls, and the expression of serum miR-320a was significantly correlated with clinical CHF indexes. miR-320a expression levels were significantly increased in exosomes isolated from patients with CHF compared with those isolated from healthy controls. Phosphoinositide-3-kinase catalytic α polypeptide gene (PIK3CA) expression levels and sST2 content were increased in HEH2 cells following transfection with miR-320a mimics compared with NC-mimic, whereas miR-320a inhibitor displayed contrasting effects by reduced the cell viability and apoptosis in myocardial fibroblasts compared with the NC-inhibitor group. The protein expression levels of collagen I, collagen III, α-smooth muscle actin, phosphorylated (p)-mTOR (ser 2448)/mTOR, p-Akt (ser 473)/Akt, p-Akt (thr 308)/Akt and PIK3CA were significantly increased in miR-320a mimic-transfected HEH2 cells compared with the NC-mimics groups. By contrast, miR-320a inhibitor notably downregulated the expression levels of these proteins compared with the NC-inhibitor group. Collectively, the results of the present study demonstrated that miR-320a promoted myocardial fibroblast proliferation via regulating the PIK3CA/Akt/mTOR signaling pathway in HEH2 cells, suggesting that serum exosomal miR-320a may serve as a potential biomarker for the diagnosis of CHF.
Collapse
Affiliation(s)
- Qing-Gao Wang
- Department of Cardiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530023, P.R. China
| | - Brian Chi-Yan Cheng
- College of Professional and Continuing Education, The Hong Kong Polytechnic University, Hong Kong 999077, SAR, P.R. China
| | - Ya-Zhou He
- Department of Cardiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530023, P.R. China
| | - Li-Juan Li
- Department of Cardiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530023, P.R. China
| | - Yun Ling
- School of Nursing, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530200, P.R. China
| | - Gan Luo
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P.R. China
| | - Li Wang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P.R. China
| | - Shan Liang
- Department of Cardiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530023, P.R. China
| | - Yi Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P.R. China
| |
Collapse
|
31
|
Lichtenauer M, Jirak P, Paar V, Sipos B, Kopp K, Berezin AE. Heart Failure and Diabetes Mellitus: Biomarkers in Risk Stratification and Prognostication. APPLIED SCIENCES 2021; 11:4397. [DOI: 10.3390/app11104397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
Heart failure (HF) and type 2 diabetes mellitus (T2DM) have a synergistic effect on cardiovascular (CV) morbidity and mortality in patients with established CV disease (CVD). The aim of this review is to summarize the knowledge regarding the discriminative abilities of conventional and novel biomarkers in T2DM patients with established HF or at higher risk of developing HF. While conventional biomarkers, such as natriuretic peptides and high-sensitivity troponins demonstrate high predictive ability in HF with reduced ejection fraction (HFrEF), this is not the case for HF with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous disease with a high variability of CVD and conventional risk factors including T2DM, hypertension, renal disease, older age, and female sex; therefore, the extrapolation of predictive abilities of traditional biomarkers on this population is constrained. New biomarker-based approaches are disputed to be sufficient for improving risk stratification and the prediction of poor clinical outcomes in patients with HFpEF. Novel biomarkers of biomechanical stress, fibrosis, inflammation, oxidative stress, and collagen turn-over have shown potential benefits in determining prognosis in T2DM patients with HF regardless of natriuretic peptides, but their role in point-to-care and in routine practice requires elucidation in large clinical trials.
Collapse
|
32
|
Stege NM, de Boer RA, van den Berg MP, Silljé HHW. The Time Has Come to Explore Plasma Biomarkers in Genetic Cardiomyopathies. Int J Mol Sci 2021; 22:2955. [PMID: 33799487 PMCID: PMC7998409 DOI: 10.3390/ijms22062955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
For patients with hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) or arrhythmogenic cardiomyopathy (ACM), screening for pathogenic variants has become standard clinical practice. Genetic cascade screening also allows the identification of relatives that carry the same mutation as the proband, but disease onset and severity in mutation carriers often remains uncertain. Early detection of disease onset may allow timely treatment before irreversible changes are present. Although plasma biomarkers may aid in the prediction of disease onset, monitoring relies predominantly on identifying early clinical symptoms, on imaging techniques like echocardiography (Echo) and cardiac magnetic resonance imaging (CMR), and on (ambulatory) electrocardiography (electrocardiograms (ECGs)). In contrast to most other cardiac diseases, which are explained by a combination of risk factors and comorbidities, genetic cardiomyopathies have a clear primary genetically defined cardiac background. Cardiomyopathy cohorts could therefore have excellent value in biomarker studies and in distinguishing biomarkers related to the primary cardiac disease from those related to extracardiac, secondary organ dysfunction. Despite this advantage, biomarker investigations in cardiomyopathies are still limited, most likely due to the limited number of carriers in the past. Here, we discuss not only the potential use of established plasma biomarkers, including natriuretic peptides and troponins, but also the use of novel biomarkers, such as cardiac autoantibodies in genetic cardiomyopathy, and discuss how we can gauge biomarker studies in cardiomyopathy cohorts for heart failure at large.
Collapse
Affiliation(s)
| | | | | | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, AB43, 9713 GZ Groningen, The Netherlands; (N.M.S.); (R.A.d.B.); (M.P.v.d.B.)
| |
Collapse
|
33
|
Miftode RS, Petriș AO, Onofrei Aursulesei V, Cianga C, Costache II, Mitu O, Miftode IL, Șerban IL. The Novel Perspectives Opened by ST2 in the Pandemic: A Review of Its Role in the Diagnosis and Prognosis of Patients with Heart Failure and COVID-19. Diagnostics (Basel) 2021; 11:diagnostics11020175. [PMID: 33530550 PMCID: PMC7911622 DOI: 10.3390/diagnostics11020175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/01/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
The increasing incidence of coronavirus disease 19 (COVID-19) and its polymorphic clinical manifestations due to local and systemic inflammation represent a high burden for many public health systems. Multiple evidence revealed the interdependence between the presence of cardiovascular comorbidities and a severe course of COVID-19, with heart failure (HF) being incriminated as an independent predictor of mortality. Suppression of tumorigenicity-2 ST2 has emerged as one of the most promising biomarkers in assessing the evolution and prognosis of patients with HF. The uniqueness of ST2 is determined by its structural particularities. Its transmembrane isoform exerts cardioprotective effects, while the soluble isoform (sST2), which is detectable in serum, is associated with myocardial fibrosis and poor outcome in patients with HF. Some recent data also suggested the potential role of sST2 as a marker of inflammation, while other studies highlighted it as a valuable prognostic factor in patients with COVID-19. In this review, we summarized the pathways by which sST2 is related to myocardial injury and its connection to the severity of inflammation in patients with COVID-19. Also, we reviewed possible perspectives of using it as a dual cardio-inflammatory biomarker, for both early diagnosis, risk stratification and prognosis assessment of patients with concomitant HF and COVID-19.
Collapse
Affiliation(s)
- Radu-Stefan Miftode
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (R.-S.M.); (A.O.P.); (I.-I.C.); (O.M.)
| | - Antoniu Octavian Petriș
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (R.-S.M.); (A.O.P.); (I.-I.C.); (O.M.)
| | - Viviana Onofrei Aursulesei
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (R.-S.M.); (A.O.P.); (I.-I.C.); (O.M.)
- Correspondence:
| | - Corina Cianga
- Department of Immunology, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Irina-Iuliana Costache
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (R.-S.M.); (A.O.P.); (I.-I.C.); (O.M.)
| | - Ovidiu Mitu
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (R.-S.M.); (A.O.P.); (I.-I.C.); (O.M.)
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Ionela-Lăcrămioara Șerban
- Department of Morpho-Functional Sciences (II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| |
Collapse
|
34
|
Kuster N, Huet F, Dupuy AM, Akodad M, Battistella P, Agullo A, Leclercq F, Kalmanovich E, Meilhac A, Aguilhon S, Cristol JP, Roubille F. Multimarker approach including CRP, sST2 and GDF-15 for prognostic stratification in stable heart failure. ESC Heart Fail 2020; 7:2230-2239. [PMID: 32649062 PMCID: PMC7524044 DOI: 10.1002/ehf2.12680] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Aims Inflammation and cardiac remodelling are common and synergistic pathways in heart failure (HF). Emerging biomarkers such as soluble suppression of tumorigenicity 2 (sST2) and growth differentiation factor‐15 (GDF‐15), which are linked to inflammation and fibrosis process, have been proposed as prognosis factors. However, their potential additive values remain poorly investigated. Methods and results Here, we aimed at evaluating inflammatory and remodelling biomarkers to predict both short‐term and long‐term mortality in a population with chronic HF in comparison with other classical clinical or biological markers (i.e. N terminal pro brain natriuretic peptide, hs‐cTnT, C‐reactive protein) alone or using meta‐analysis global group in chronic HF risk score in a cohort of 182 patients followed during 80 months (interquartile range: 12.3–90.0). Proportional hazard assumption does not hold for sST2 and C‐reactive protein, and follow‐up was split into short term (less than 1 year), midterm (between 1 and 5 years), and long term (after 5 years). In univariate analysis, C‐reactive protein and sST2 were predictive of short‐term mortality but not of middle term and long term whereas GDF‐15 was predictive of short and mid‐term but not of long‐term mortality. In a multivariate model after adjustment for meta‐analysis global group in chronic HF score including the three markers, only sST2 was predictive of short‐term mortality (P = 0.0225), and only GDF‐15 was predictive of middle term mortality (P = 0.0375). None of the markers was predictive of long‐term mortality. Conclusions Our results demonstrate that both sST2 and GDF‐15 significantly improve the prognosis evaluation of HF patients and suggest that the value of GDF‐15 is more sustained overtime and could predict middle term events.
Collapse
Affiliation(s)
- Nils Kuster
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Anne-Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France
| | - Mariama Akodad
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Pascal Battistella
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Audrey Agullo
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Florence Leclercq
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Eran Kalmanovich
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Alexandra Meilhac
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Francois Roubille
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| |
Collapse
|
35
|
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
|
36
|
Novel Molecular Mechanisms of Pulmonary Hypertension: A Search for Biomarkers and Novel Drug Targets-From Bench to Bed Site. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7265487. [PMID: 32566097 PMCID: PMC7261339 DOI: 10.1155/2020/7265487] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022]
Abstract
Pulmonary hypertension (PH) is defined as increased mean pulmonary artery pressure (mPAP) above 25 mmHg, measured at rest by right heart catheterization. The exact global prevalence of PH is difficult to estimate, mainly due to the complex aetiology, and its spread may be underestimated. To date, numerous studies on the aetiology and pathophysiology of PH at molecular level were conducted. Simultaneously, some clinical studies have shown potential usefulness of well-known and widely recognized cardiovascular biomarkers, but their potential clinical usefulness in diagnosis and management of PH is poor due to their low specificity accompanied with numerous other cardiovascular comorbidities of PH subjects. On the other hand, a large body of basic research-based studies provides us with novel molecular pathomechanisms, biomarkers, and drug targets, according to the evidence-based medicine principles. Unfortunately, the simple implementation of these results to clinical practice is impossible due to a large heterogeneity of the PH pathophysiology, where the clinical symptoms constitute only a common denominator and a final result of numerous crosstalking metabolic pathways. Therefore, future studies, based mostly on translational medicine, are needed in order to both organize better the pathophysiological classification of various forms of PH and define precisely the optimal diagnostic markers and therapeutic targets in particular forms of PH. This review paper summarizes the current state of the art regarding the molecular background of PH with respect to its current classification. Novel therapeutic strategies and potential biomarkers are discussed with respect to their limitations in use in common clinical practice.
Collapse
|