1
|
Monzo L, Kobayashi M, Ferreira JP, Lamiral Z, Delles C, Clark AL, Edelmann F, González A, Heymans S, Pellicori P, Petutschnigg J, Verdonschot JAJ, Rossignol P, Cleland JGF, Zannad F, Girerd N. Echocardiographic and biomarker characteristics in diabetes, coronary artery disease or both: insights from HOMAGE trial. Cardiovasc Diabetol 2025; 24:111. [PMID: 40057728 PMCID: PMC11890510 DOI: 10.1186/s12933-025-02609-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Coronary artery disease (CAD) and diabetes mellitus (DM) can induce changes in myocardial structure and function, thereby increasing the risk of heart failure (HF). We aimed to identify the alterations in echocardiographic variables and circulating biomarkers associated with DM, CAD, or both and to assess the effect of spironolactone on them. METHODS The "Heart OMics in AGEing" (HOMAGE) trial evaluated the effect of spironolactone on circulating markers of fibrosis over 9 months of follow-up in people at risk for HF. From the initial population (N = 527) of the HOMAGE trial, a total of 495 participants (mean age 74 years, 25% women) were categorized according to clinical phenotype (DM-/CAD + vs. DM+/CAD- vs. DM+/CAD+), while the DM-/CAD- group was excluded due to the low sample size (N = 32). Multivariable linear regression analysis was used to assess the relations between variables and DM/CAD status. RESULTS At baseline, participants with DM, whether or not they had CAD, showed lower markers of type I collagen synthesis (procollagen type I C-terminal propeptide; β [95% CI]: DM+/CAD-: -6.973 [-13.778; -0.167]; DM+/CAD+: -9.039 [-15.174; -2.903]), reduced left ventricular volumes (β [95% CI]: end-diastolic, DM+/CAD-: -6.323 [-9.696; -2.951]; DM+/CAD+: -2.503 [-5.531; 0.526]; end-systolic, DM+/CAD-: -2.905 [-4.817; -0.992]; DM+/CAD+: -1.400 [-3.120; 0.320]) and higher levels of galectin-3 (Exponential β [95% CI]: DM+/CAD-: 1.127 [1.050; 1.209]; DM+/CAD+: 1.118 [1.048; 1.192]), and growth differentiation factor-15 (Exponential β [95% CI]: DM+/CAD-: 1.542 [1.360; 1.747]; DM+/CAD+: 1.535 [1.370; 1.720]), along with an elevated E/e' ratio (β [95% CI]: DM+/CAD-: 1.355 [0.462; 2.248]; DM+/CAD+: 0.879 [0.067; 1.690]), compared with DM-/CAD + individuals (all p < 0.05). At follow-up, the effect of spironolactone on echocardiographic variables and circulating biomarkers was not significantly different across DM/CAD phenotypes (all p-interaction > 0.05), except for a more pronounced reduction in GDF-15 in the DM+/CAD + group at the 1-month visit (p-interaction = 0.03). CONCLUSIONS Among HOMAGE trial participants, diabetes was a powerful driver of biomarker and echocardiographic alterations irrespectively of CAD. These alterations were mainly related to the domains of inflammation and diastolic function.
Collapse
Affiliation(s)
- Luca Monzo
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France.
| | - Masatake Kobayashi
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
- Department of Cardiology, Tokyo Medical University Hospital, Tokyo, Japan
| | - João Pedro Ferreira
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
- Department of Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Zohra Lamiral
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew L Clark
- Department of Cardiology, Castle Hill Hospital, University of Hull, East Riding of Yorkshire, UK
| | - Frank Edelmann
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie & Intensivmedizin, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Arantxa González
- Department of Pathology, Anatomy and Physiology Universidad de Navarra and IdiSNA, CIMA Universidad de Navarra, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pierpaolo Pellicori
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Johannes Petutschnigg
- Deutsches Herzzentrum der Charité, Klinik für Kardiologie, Angiologie & Intensivmedizin, Augustenburger Platz 1, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Job A J Verdonschot
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Rossignol
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
- Department of Medicine and Nephrology-Hemodialysis, Princess Grace Hospital, Monaco, Monaco
- M-CRIN (Monaco Clinical Research Infrastructure Network), Monaco, Monaco
- Monaco Private Hemodialysis Centre, Monaco, Monaco
| | - John G F Cleland
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France
| | - Nicolas Girerd
- Centre d'Investigations Cliniques Plurithématique 1433 and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, Nancy, France.
| |
Collapse
|
2
|
Wimalanathan T, Paus MF, Brox Skranes J, Berge T, Tveit A, Røsjø H, Omland T, Lyngbakken MN, Lagethon Heck S. Associations between Growth Differentiation Factor 15, Cardiac Troponin T, and N-terminal pro-B-type Natriuretic Peptide, and Future Myocardial Fibrosis Assessed by Cardiac Magnetic Resonance Imaging: Data from the Akershus Cardiac Examination 1950 Study. J Appl Lab Med 2025; 10:392-405. [PMID: 39707823 DOI: 10.1093/jalm/jfae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Myocardial fibrosis is associated with a poor outcome for patients with cardiovascular disease (CVD). Growth differentiation factor 15 (GDF-15) concentrations predict the risk of death in patients with CVD, but the underlying pathophysiological mechanisms are poorly understood. We aimed to assess the associations between biomarkers of cellular stress and inflammation (GDF-15), cardiac injury (cardiac troponin T [cTnT]), and stretch (N-terminal pro-B-type natriuretic peptide [NT-proBNP]), and subsequent focal and diffuse myocardial fibrosis assessed by cardiac magnetic resonance (CMR) imaging. METHODS We measured GDF-15, cTnT, and NT-proBNP in 200 study participants without known coronary artery disease or renal dysfunction from the population-based Akershus Cardiac Examination 1950 Study at baseline in 2012 to 2015. Focal myocardial scars and diffuse fibrosis were assessed by late gadolinium enhancement imaging and septal extracellular volume fraction (ECV) by CMR 4 to 7 years later. The relationships between cardiac biomarkers and CMR parameters were assessed by logistic regression analysis adjusted for common cardiovascular risk factors. RESULTS The median age was 63.9 (interquartile range 63.4-64.5) years and 49% were women. GDF-15 (adjusted odds ratio [aOR] 4.40, 95% CI 1.09-17.72) and cTnT (aOR 1.59, 95% CI 1.01-2.50) were associated with nonischemic scars in the fully adjusted model. cTnT (aOR 2.45, 95% CI 1.41-4.25) and NT-proBNP (aOR 3.12, 95% CI 1.55-6.28) were associated with ischemic scars. None of the biomarkers were significantly associated with elevated ECV. CONCLUSIONS In a general population cohort, GDF-15, an emerging biomarker of cellular stress and inflammation, associates with nonischemic scars. Biomarkers of myocardial injury and stretch associate with ischemic scars, while no biomarker was associated with diffuse fibrosis as assessed by CMR.
Collapse
Affiliation(s)
- Thakshani Wimalanathan
- Department of Diagnostic Imaging, Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Michael Fredrik Paus
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Julia Brox Skranes
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trygve Berge
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arnljot Tveit
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Helge Røsjø
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Akershus Clinical Research Center (ACR), Division of Research and Innovation, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - M N Lyngbakken
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Siri Lagethon Heck
- Department of Diagnostic Imaging, Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
3
|
Chen YL, You J, Guo Y, Zhang Y, Yao BR, Wang JJ, Chen SD, Ge YJ, Yang L, Wu XR, Wu BS, Zhang YR, Dong Q, Feng JF, Tian M, Cheng W, Yu JT. Identifying proteins and pathways associated with multimorbidity in 53,026 adults. Metabolism 2025; 164:156126. [PMID: 39740741 DOI: 10.1016/j.metabol.2024.156126] [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: 10/13/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND AND AIMS Multimorbidity, the coexistence of multiple chronic diseases, is a rapidly expanding global health challenge, carrying profound implications for patients, caregivers, healthcare systems, and society. Investigating the determinants and drivers underlying multiple chronic diseases is a priority for disease management and prevention. METHOD This prospective cohort study analyzed data from the 53,026 participants in the UK Biobank from baseline (2006 to 2010) across 13.3 years of follow-up. Using Cox proportional hazards regression model, we characterized shared and unique associations across 38 incident outcomes (31 chronic diseases, 6 system mortality and all-cause mortality). Furthermore, ordinal regression models were used to assess the association between protein levels and multimorbidity (0-1, 2, 3-4, or ≥ 5 chronic diseases). Functional and tissue enrichment analysis were employed for multimorbidity-associated proteins. The upstream regulators of above proteins were identified. RESULTS We demonstrated 972 (33.3 %) proteins were shared across at least two incident chronic diseases after Bonferroni correction (P < 3.42 × 10-7, 93.3 % of those had consistent effects directions), while 345 (11.8 %) proteins were uniquely linked to a single chronic disease. Remarkably, GDF15, PLAUR, WFDC2 and AREG were positively associated with 20-24 incident chronic diseases (hazards ratios: 1.21-3.77) and showed strong associations with multimorbidity (odds ratios: 1.33-1.89). We further identified that protein levels are explained by common risk factors, especially renal function, liver function, inflammation, and obesity, providing potential intervention targets. Pathway analysis has underscored the pivotal role of the immune response, with the top three transcription factors associated with proteomics being NFKB1, JUN and RELA. CONCLUSIONS Our results enhance the understanding of the biological basis underlying multimorbidity, offering biomarkers for disease identification and novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jia You
- Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bing-Ran Yao
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | | | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Mei Tian
- Huashan Hospital & Human Phenome Institute, Fudan University, Shanghai, China; Department of Nuclear Medicine/PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Weng Y, Zhou T, Ye H. Development and assessment of a mortality risk prediction nomogram model for pneumocystis disease in ICU within 28 days. Sci Rep 2025; 15:2410. [PMID: 39827208 PMCID: PMC11742898 DOI: 10.1038/s41598-025-86696-3] [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: 10/31/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
To develop and assess a nomogram predictive model for evaluating the 28-day mortality risk in patients diagnosed with Pneumocystis who have been admitted to the intensive care unit (ICU). From 2008 to 2022, clinical data on patients with Pneumocystis were collected using the American Critical Care Medical Information Database IV (MIMIC-IV). Initially, 63 significant predictive indicators were included, with ICU admission as the time node and all-cause mortality within 28 days as the outcome. Using complete data modeling, the variable selection approach combines two methods: Lasso regression (glmnet package) and collinearity screening (car package). Use the bootstrap method 1000 times for internal validation. Calculate the AUC, mean sensitivity, and specificity of 1000 resamplings, as well as the 95% confidence interval, and then plot the ROC, calibration, and DCA curves. The patients were split into two groups based on their 28-day survival status: 83 cases (67.48%) in the survival group and 40 instances (32.52%) in the death group. Five variables-the history of malignant tumors, Lods scores, Oasis scores, and complications of shock and severe renal injury-were eventually included in the entire sample after screening. According to Receiver Operating Characteristic (ROC) analysis, the model's sensitivity was 0.600 (95% CI 0.448-0.752), specificity was 0.904 (95% CI 0.840-0.967), and AUC was 0.814 (95% CI 0.732-0.897). The DCA curve indicates that the model application has high accuracy, which leads to a net benefit for population prediction, and the model calibration curve demonstrates good calibration accuracy. The bootstrap model's 1000 internal validation results demonstrate that the model's calibration performance is good and that its accuracy is highest when the probability of patient outcome events falls between 35 and 60%, which yields the highest net benefit for population prediction. This study developed a nomogram utilizing MIMIC-IV clinical big data to predict the 28-day mortality risk in patients with Pneumocystis disease. Incorporating five critical factors, the nomogram offers a user-friendly, visual method for calculating personalized risk scores based on patient-specific information, including medical history, laboratory results, and clinical scores. Demonstrating robust discrimination and calibration, this tool provides clinicians with a valuable resource for assessing prognosis and making evidence-based treatment decisions.
Collapse
Affiliation(s)
- Yiru Weng
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, People's Republic of China
| | - Tingting Zhou
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, People's Republic of China
| | - Honghua Ye
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, People's Republic of China.
| |
Collapse
|
5
|
Huang S, Yu LJ, Sun GF, Zhang ZX. Short-term predictive value of sST2 in patients with STEMI following primary PCI: a prospective observational study. BMC Cardiovasc Disord 2025; 25:21. [PMID: 39819309 PMCID: PMC11737151 DOI: 10.1186/s12872-025-04488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
AIM The objective of this study was to investigate the level of soluble suppression of tumorigenicity-2 (sST2) in patients with acute ST-segment elevation myocardial infarction (STEMI) following primary percutaneous coronary intervention (PCI), and to provide a new biomarker for clinical management and prognosis assessment. METHOD This was a prospective study. 148 STEMI patients following primary PCI were enrolled and divided into 2 groups by the median value of sST2 and afterwards followed up for 30 days to access the occurrence of major adverse cardiac events (MACEs), which were defined as cardiovascular death, heart failure and recurrent MI. RESULTS sST2 ranged from 20.57 to 98.96 ng/mL. High sST2 group had higher MACEs rate compared to low sST2 group (28.8% vs. 8.0%, P = 0.001). sST2 was positively correlated with age (r = 0.181, P = 0.027), SYNTAX score (r = 0.257, P = 0.002), high-sensitive C-reactive protein (hs-CRP) (r = 0.225, P = 0.006), B-type natriuretic peptide (BNP) (r = 0.225, P = 0.006) and negatively with left ventricle ejection fraction (LVEF) (r = -0.197, P = 0.016). After adjustment for clinical variables, sST2 level (OR 3.680, P = 0.015) and LVEF (OR 0.880, P < 0.001) remained independent predictors of 30-days MACEs. In receiver operating characteristic curve (ROC) analyses, the area under the curve (AUC) of sST2 for predicting 30-days MACEs was 0.755(P < 0.001). The AUC of sST2 combining hs-CRP and LVEF for prediction was 0.828(95%CI [0.743-0.912], P < 0.001). CONCLUSION sST2 level after primary PCI was an independent risk factor of MACEs in STEMI patients through 30 days follow-up.
Collapse
Affiliation(s)
- Shan Huang
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Huli District, Xiamen, China
| | - Lu-Jiao Yu
- Department of Geriatrics, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China
| | - Guang-Feng Sun
- Department of Emergency, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Xiamen, China
| | - Zi-Xin Zhang
- Department of Cardiology, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China.
| |
Collapse
|
6
|
Meng Q, Gan X, Zu C, Zhang Y, He P, Ye Z, Su X, Wei Y, Qin X. Six-year change in high-sensitivity cardiac troponin T with subsequent stroke risk in the general population. J Epidemiol Community Health 2025; 79:138-145. [PMID: 39304191 DOI: 10.1136/jech-2024-222517] [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: 05/22/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The association between change in high-sensitivity cardiac troponin T (hs-cTnT) and stroke risk in the general population remains unknown. We aimed to assess the association of a 6-year change in hs-cTnT with incident stroke and its subtypes in the general American adult population. METHODS 8675 middle-aged adults without prevalent cardiovascular disease from the Atherosclerosis Risk in Communities study were included. Hs-cTnT was measured at two time points (visits 2 and 4), 6 years apart. The relative percentage change of hs-cTnT was defined as hs-cTnT at visit 4 minus that at visit 2, divided by hs-cTnT at visit 2. The study outcome was incident stroke and its subtypes. All data were analysed in 2023. RESULTS Over a median follow-up of 20.1 years, 682 incident strokes occurred, including 593 ischaemic and 89 haemorrhagic strokes. For absolute change, using low/low group as reference category, the low/high (adjusted HR 1.44, 95% CI 1.03 to 2.02) and high/high (adjusted HR 1.47, 95% CI 0.93 to 2.34) groups were associated with higher risk of stroke. Moreover, the relative percentage change in hs-cTnT with stroke followed an inverted L-shaped association, levelling off at about 75% increase in hs-cTnT (P for nonlinearity=0.009). Compared with those with ≤50% change in hs-cTnT, participants with >50% increase in hs-cTnT had a higher risk of stroke (adjusted HR 1.30, 95% CI 1.03 to 1.64). Similar results were found for ischaemic stroke. No significant association was found for haemorrhagic stroke. CONCLUSION Temporal increase in hs-cTnT was associated with a higher risk of incident total and ischaemic stroke in the general population.
Collapse
Affiliation(s)
- Qiguo Meng
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Institute of Biomedicine, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoqin Gan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
| | - Cheng Zu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Institute of Biomedicine, Anhui Medical University, Hefei, Anhui, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
| | - Xinyue Su
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Institute of Biomedicine, Anhui Medical University, Hefei, Anhui, China
| | - Yuanxiu Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Institute of Biomedicine, Anhui Medical University, Hefei, Anhui, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Guangzhou, Guangdong, China
- National Clinical Research Center for Kidney Disease, Guangzhou, Guangdong, China
- Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Institute of Biomedicine, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
Myrmel GMS, Wereski R, Karaji I, Saeed N, Aakre KM, Mills NL, Pedersen ER. Cardiac biomarkers and CT coronary angiography for the assessment of coronary heart disease. Clin Biochem 2025; 135:110857. [PMID: 39586418 DOI: 10.1016/j.clinbiochem.2024.110857] [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: 09/23/2024] [Revised: 10/23/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Over the last 30 years, the widespread use of cardiac biomarkers has transformed the diagnostic evaluation of patients with coronary heart disease. Cardiac troponin is integral to the definition of acute myocardial infarction. High-sensitivity cardiac troponin (hs-cTn) assays can improve risk stratification to facilitate both the rapid rule out of myocardial infarction and prediction of future cardiovascular events. Numerous circulating biomarkers representing different pathological pathways improve prediction of atherosclerotic cardiovascular disease (ACVD) and coronary artery disease (CAD). In parallel, coronary computed tomography angiography (CCTA) has become the most widely used imaging modality for the evaluation of patients with possible angina. CCTA now allows for the quantification of coronary calcification, atherosclerotic plaque volume and different plaque characteristics, enabling the identification high-risk features and inflammation. In the future, the use of CCTA is likely to extend to risk stratification for the prevention of ACVD. As such, how to integrate these diagnostic and prognostic circulating and imaging biomarkers is a topic of considerable interest. This review aims to describe current status and future possibilities for the integration of CCTA and cardiac biomarker testing to improve the identification and treatment of individuals with coronary heart disease and heightened cardiovascular risk.
Collapse
Affiliation(s)
| | - Ryan Wereski
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Iman Karaji
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Nasir Saeed
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin Moberg Aakre
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Eva Ringdal Pedersen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Skau E, Wagner P, Leppert J, Ärnlöv J, Hedberg P. Determinants of growth differentiation factor 15 plasma levels in outpatients with peripheral arterial disease. Ups J Med Sci 2024; 129:11001. [PMID: 39780955 PMCID: PMC11708457 DOI: 10.48101/ujms.v129.11001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/27/2024] [Accepted: 08/27/2024] [Indexed: 01/11/2025] Open
Abstract
Background Growth differentiation factor 15 (GDF-15) is a robust prognostic biomarker in patients with cardiovascular (CV) disease, and a better understanding of its clinical determinants is desirable. We aimed to study the associations between GDF-15 levels and traditional CV risk factors, indicators of atherosclerotic burden, and cardiac geometry and dysfunction in outpatients with peripheral arterial disease (PAD). Methods An explorative cross-sectional study (Study of Atherosclerosis in Vastmanland, Västerås, Sweden) included 439 outpatients with carotid or lower extremity PAD. The mean age was 70 years (standard deviation [SD] 7), and 59% of the patients were men. Plasma levels of GDF-15 were obtained along with potential determinants, including medical history, biochemical data, echocardiographic measures of cardiac geometry and function, ankle-brachial index (ABI), and carotid ultrasonographic data on intima-media thickness (IMT) and occurrence of carotid stenosis. The relations between GDF-15 concentrations (transformed with the natural logarithm) and the different determinants were evaluated using uni- and multivariable linear regression models. All pre-specified variables were included in the multivariable models. Results The multivariable analysis identified independent relations of GDF-15 with several of the included variables (adjusted R 2 = 0.48). Diabetes (beta coefficient [β] of 0.37, 95% confidence interval [95% CI] 0.25 to 0.50), low-density lipoprotein (LDL) cholesterol (β = -0.22, 95% confidence interval [CI]: -0.34 to -0.09), and physical activity (β = -0.16, 95% CI: -0.25 to -0.06) had the strongest associations. In contrast, no significant independent associations with GDF-15 level were observed for cardiac geometry and function, ABI, IMT, or carotid stenosis. Conclusions Circulating GDF-15 is more strongly associated with traditional CV risk factors, especially diabetes, LDL cholesterol, and physical activity than with specific indicators of atherosclerotic burden or cardiac dysfunction. To better understand the pathophysiological role of GDF-15 and its link to clinical outcomes in patients with PAD, future studies should focus on the metabolic processes involved in atherosclerotic disease.
Collapse
Affiliation(s)
- Emma Skau
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Cardiology, Danderyd University Hospital, Stockholm, Sweden
| | - Philippe Wagner
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Clinical Physiology, Västmanland County Hospital, Västerås, Sweden
| |
Collapse
|
9
|
Kosyakovsky LB, de Boer RA, Ho JE. Screening for Heart Failure: Biomarkers to Detect Heightened Risk in the General Population. Curr Heart Fail Rep 2024; 21:591-603. [PMID: 39287754 DOI: 10.1007/s11897-024-00686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW Heart failure (HF) represents a growing global burden of morbidity and mortality. Identifying individuals at risk for HF development is increasingly important, particularly given the advent of disease-modifying therapies for HF as well as its major risk factors such as obesity actalnd diabetes. We aim to review the key circulating biomarkers associated with future HF which may contribute to HF risk prediction. RECENT FINDINGS While current guidelines recommend the use of natriuretic peptides and cardiac troponins in HF risk stratification, there are a diverse array of other emerging protein, metabolic, transcriptomic, and genomic biomarkers of future HF development. These biomarkers not only lend insight into the underlying pathophysiology of HF, which spans inflammation to cardiac fibrosis, but also offer an opportunity to further refine HF risk in addition to established biomarkers. As evolving techniques in molecular biology enable an increased understanding of the complex biologic contributions to HF pathophysiology, there is an important opportunity to construct integrated clinical and multi-omic models to best capture HF risk. Moving forward, future studies should seek to understand the contributions of sex differences, underlying comorbidity burden, and HF subtypes to an individual's HF risk. Further studies are necessary to fully define the clinical utility of biomarker screening approaches to refine HF risk assessment, as well as to link risk assessment directly to preventive strategies for HF.
Collapse
Affiliation(s)
- Leah B Kosyakovsky
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA
| | - Rudolf A de Boer
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, the Netherlands
| | - Jennifer E Ho
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA.
| |
Collapse
|
10
|
Hsieh MJ, Yeh JK, Huang YC, Ho MY, Chen DY, Lee CH, Wang CY, Chang SH, Chen CC, Hsieh IC. Cardiac power output associated with hospitalization and mortality in coronary artery disease patients at stage B heart failure. IJC HEART & VASCULATURE 2024; 55:101521. [PMID: 39430397 PMCID: PMC11490734 DOI: 10.1016/j.ijcha.2024.101521] [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: 07/09/2024] [Revised: 09/07/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024]
Abstract
Background Cardiac power output (CPO) predicts outcomes in advanced heart failure (HF) and cardiogenic shock, but its role in early HF stages is unclear. This study assessed the prognostic value of CPO in coronary artery disease patients with asymptomatic left ventricular systolic dysfunction (ALVSD) at stage B HF. Methods We conducted a retrospective analysis of coronary artery disease patients who underwent coronary and pulmonary artery catheterization between 2006 and 2016. Stage B HF with ALVSD was defined as left ventricular ejection fraction < 50 %, without HF symptoms, signs, or prior HF hospitalization. CPO was derived from invasive hemodynamic parameters. Endpoints included HF hospitalization, cardiovascular mortality, and all-cause mortality over a 5-year follow-up. Results A total of 783 coronary artery disease patients with ALVSD at stage B HF were enrolled. Incidence rates (per 1000 person-years) were 13.9 for HF hospitalization, 14.5 for cardiovascular mortality, and 23.7 for all-cause mortality.Multivariate analysis adjusting for covariates demonstrated that CPO was independent associated with all endpoints. Patients with a low CPO (<0.97 Watts) were at significantly higher risk for HF hospitalization (adjusted hazard ratio [HR]: 4.04; 95 % CI: 1.53 - 10.6; p = 0.005), cardiovascular mortality (adjusted HR: 2.73; 95 % CI: 1.19 - 6.27; p = 0.018), and all-cause mortality (adjusted HR: 1.86; 95 % CI: 1.05 - 3.30; p = 0.035) compared to those with higher CPO, regardless of subgroup classification. Conclusion Resting CPO in patients with ALVSD is significantly associated with adverse events, including HF hospitalization and mortality, highlighting its value in early-stage HF management.
Collapse
Affiliation(s)
- Ming-Jer Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jih-Kai Yeh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chang Huang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Yun Ho
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Dong-Yi Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chao-Yung Wang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Chi Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - I-Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
11
|
Lorenz EC, Smith BH, Liang Y, Park WD, Bentall AJ, Dhala AF, Waterman AD, Kennedy CC, Hickson LJ, Rule AD, Cheville AL, LeBrasseur NK, Stegall MD. Increased Pretransplant Inflammatory Biomarkers Predict Death With Function After Kidney Transplantation. Transplantation 2024; 108:2434-2445. [PMID: 38913783 PMCID: PMC11666810 DOI: 10.1097/tp.0000000000005103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Chronic systemic inflammation is associated with mortality in patients with chronic kidney disease, cardiovascular disease, and diabetes. The goal of this study was to examine the relationship between pretransplant inflammatory biomarkers (growth differentiation factor-15 [GDF-15], interleukin-6 [IL-6], soluble tumor necrosis factor receptor-1, monokine induced by gamma interferon/chemokine [C-X-C motif] ligand 9 [MIG/CXCL9], monocyte chemoattractant protein-1, soluble FAS, tumor necrosis factor-α, interleukin-15, and interleukin-1β) and death with function (DWF) after kidney transplantation (KT). METHODS We retrospectively measured inflammatory biomarker levels in serum collected up to 1 y before KT (time from blood draw to KT was 130 ± 110 d) in recipients transplanted between January 2006 and December 2018. Kaplan-Meier estimation, Cox regression, and Gradient Boosting Machine modeling were used to examine the relationship between inflammatory biomarkers and DWF. RESULTS Our cohort consisted of 1595 KT recipients, of whom 62.9% were male and 83.2% were non-Hispanic White. Over a mean follow-up of 7.4 ± 3.9 y, 21.2% of patients (n = 338) experienced DWF. Patients with the highest quartile levels of GDF-15 (>4766 pg/mL), IL-6 (>6.11 pg/mL), and MIG/CXCL9 (> 5835 pg/mL) had increased rates of DWF, and each predicted mortality independently of the others. When adjusted for clinical factors (age, diabetes, etc), the highest quartile levels of GDF-15 and IL-6 remained independently associated with DWF. Adding inflammatory markers to a clinical Cox model improved the C-statistic for DWF from 0.727 to 0.762 using a Gradient Boosting Machine modeling approach. CONCLUSIONS These findings suggest that pre-KT serum concentrations of GDF-15, IL-6, and MIG/CXCL9 may help to risk stratify and manage patients undergoing KT and suggests that chronic inflammation may play a role in mortality in KT recipients.
Collapse
Affiliation(s)
| | - Byron H Smith
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Yun Liang
- Department of Surgery, Mayo Clinic, Rochester, MN
| | | | - Andrew J Bentall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Atiya F Dhala
- Department of Surgery, Houston Methodist Hospital, Houston, TX
| | - Amy D Waterman
- Department of Surgery, Houston Methodist Hospital, Houston, TX
| | - Cassie C Kennedy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mayo Clinic, Rochester, MN
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Andrea L Cheville
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
12
|
Bellos I, Marinaki S, Lagiou P, Benetou V. Association of soluble suppression of tumorigenicity 2 with mortality and adverse outcomes in chronic kidney disease: a systematic review and meta-analysis. Clin Exp Nephrol 2024; 28:988-1003. [PMID: 38678167 PMCID: PMC11493800 DOI: 10.1007/s10157-024-02506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Early risk stratification is necessary to prevent chronic kidney disease progression and complications. This systematic review aims to evaluate the association of soluble suppression of tumorigenicity 2 (sST2), a member of the interleukin-1 receptor family, with all-cause mortality, cardiovascular disease and renal function deterioration among chronic kidney disease patients. METHODS PubMed, Scopus, Web of Science, CENTRAL and Google Scholar were systematically searched from inception to December 20, 2023. Cohort studies examining the prognostic role of sST2 levels in pre-dialysis and dialysis patients were included. In case of 3 or more studies per outcome, conventional and dose-response meta-analyses were conducted. RESULTS Overall, 21 studies were included comprising 15,100 patients. In pre-dialysis patients, the qualitative synthesis of studies suggested that high sST2 is associated with significantly increased all-cause mortality, while evidence regarding cardiovascular events or kidney disease progression was conflicting. In the dialysis population, high sST2 was linked to an elevated risk of all-cause (Hazard ratio-HR: 3.00, 95% confidence intervals-CI: 1.95-4.61) and cardiovascular (HR: 2.38, 95% CI: 1.69-3.34) mortality. Dose-response meta-analysis suggested a log-linear association of sST2 with both all-cause (χ2: 34.65, p value < 0.001) and cardiovascular (χ2: 29.14, p value < 0.001) mortality, whereas findings regarding cardiovascular events were limited with mixed results. CONCLUSIONS High sST2 values are associated with an increased risk of all-cause mortality in pre-dialysis and dialysis patients, as well as with an elevated risk of cardiovascular mortality in the dialysis population. Further studies are needed to elucidate its potential association with cardiovascular events and kidney disease progression.
Collapse
Affiliation(s)
- Ioannis Bellos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece.
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Smaragdi Marinaki
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| | - Vassiliki Benetou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| |
Collapse
|
13
|
Lyu L, Xv C, Xu J, Liu Z, He Y, Zhu W, Lin L, Yang Q, Wei Y, Wang J, Huang T, Hao B, Liu H. Growth differentiation factor-15 predicts all-cause death and major adverse cardiovascular events in patients with coronary heart disease: a prospective cohort study. J Thromb Thrombolysis 2024; 57:1109-1121. [PMID: 39068629 DOI: 10.1007/s11239-024-03019-5] [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] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
The prognostic value of growth differentiation factor-15 (GDF-15) in predicting long-term adverse outcomes in coronary heart disease (CHD) patients remains limited. Our study examines the association between GDF-15 and adverse outcomes over an extended period in CHD patients and firstly assesses the incremental prognostic effect of incorporating GDF-15 into the Framingham risk score (FRS)-based model. This single-center prospective cohort study included 3,321 patients with CHD categorized into 2,479 acute coronary syndrome (ACS) (74.6%) and 842 non-ACS (25.4%) groups. The median age was 61.0 years (range: 53.0-70.0), and 917 (27.6%) were females. Mortality and major adverse cardiovascular events (MACEs) included cardiovascular mortality, myocardial infarction (MI), stroke, and heart failure (HF) (inclusive of HF episodes requiring outpatient treatment and/or hospital admission). Cox regression models assessed the associations between GDF-15 and the incidence of all-cause mortality and MACEs. Patients were stratified into three groups based on GDF-15 levels: the first tertile group (< 1,370 ng/L), the second tertile group (1,370-2,556 ng/L), and the third tertile group (> 2,556 ng/L). The C-index, integrated discrimination improvement (IDI), net reclassification improvement (NRI), and decision curve analysis (DCA) were used to assess incremental value. Over a median 9.4-year follow-up, 759 patients (22.9%) died, and 1,291 (38.9%) experienced MACEs. The multivariate Cox model indicated that GDF-15 was significantly associated with all-cause mortality (per ln unit increase, HR = 1.49, 95% CI: 1.36-1.64) and MACEs (per ln unit increase, HR = 1.29, 95% CI: 1.20-1.38). These associations persisted when GDF-15 was analyzed as an ordinal variable (p for trend < 0.05). Subgroup analysis of ACS and non-ACS for the components of MACEs separately showed a significant association between GDF-15 and both cardiovascular mortality and HF, but no association was observed between GDF-15 and MI /stroke in both ACS and non-ACS patients. The addition of GDF-15 to the FRS-based model enhanced the discrimination for both all-cause mortality (∆ C-index = 0.009, 95% CI: 0.005-0.014; IDI = 0.030, 95% CI: 0.015-0.047; continuous NRI = 0.631, 95% CI: 0.569-0.652) and MACEs (∆ C-index = 0.009, 95% CI: 0.006-0.012; IDI = 0.026, 95% CI: 0.009-0.042; continuous NRI = 0.593, 95% CI: 0.478-0.682). DCA suggested that incorporating GDF-15 into the FRS-based model demonstrated higher net benefits compared to FRS-based models alone (All-cause mortality: FRS-based model: area under the curve of DCA (AUDC) = 0.0903, FRS-based model + GDF-15: AUDC = 0.0908; MACEs: FRS-based model: AUDC = 0.1806, FRS-based model + GDF-15: AUDC = 0.1833). GDF-15 significantly associates with the long-term prognosis of all-cause mortality and MACEs in CHD patients and significantly improves the prognostic accuracy of the FRS-based model for both outcomes.
Collapse
Affiliation(s)
- Lyu Lyu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Cui Xv
- Department of Medical Administration, The 305 Hospital of PLA, Beijing, China
| | - Juan Xu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Zhenzhen Liu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanru He
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenjing Zhu
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Lin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Wei
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinda Wang
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Taoke Huang
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Benchuan Hao
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
- Medical School of Chinese PLA, Beijing, China.
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, China.
| |
Collapse
|
14
|
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
|
15
|
Spanos M, Gokulnath P, Li G, Hutchins E, Meechoovet B, Sheng Q, Chatterjee E, Sharma R, Carnel-Amar N, Lin C, Azzam C, Ghaeli I, Amancherla KV, Victorino JF, Garcia-Mansfield K, Pfeffer R, Sahu P, Lindman BR, Elmariah S, Gamazon ER, Betti MJ, Bledsoe X, Lance ML, Absi T, Su YR, Do N, Contreras MG, Varrias D, Kladas M, Radulovic M, Tsiachris D, Spanos A, Tsioufis K, Ellinor PT, Tucker NR, Januzzi JL, Pirrotte P, Jovanovic-Talisman T, Van Keuren-Jensen K, Shah R, Das S. Cardiomyocyte-derived circulating extracellular vesicles allow a non-invasive liquid biopsy of myocardium in health and disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.19.24314009. [PMID: 39371135 PMCID: PMC11451713 DOI: 10.1101/2024.09.19.24314009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The ability to track disease without tissue biopsy in patients is a major goal in biology and medicine. Here, we identify and characterize cardiomyocyte-derived extracellular vesicles in circulation (EVs; "cardiovesicles") through comprehensive studies of induced pluripotent stem cell-derived cardiomyocytes, genetic mouse models, and state-of-the-art mass spectrometry and low-input transcriptomics. These studies identified two markers (POPDC2, CHRNE) enriched on cardiovesicles for biotinylated antibody-based immunocapture. Captured cardiovesicles were enriched in canonical cardiomyocyte transcripts/pathways with distinct profiles based on human disease type (heart failure, myocardial infarction). In paired myocardial tissue-plasma from patients, highly expressed genes in cardiovesicles were largely cardiac-enriched (vs. "bulk" EVs, which were more organ non-specific) with high expression in myocardial tissue by single nuclear RNA-seq, largely in cardiomyocytes. These results demonstrate the first "liquid" biopsy discovery platform to interrogate cardiomyocyte states noninvasively in model systems and in human disease, allowing non-invasive characterization of cardiomyocyte biology for discovery and therapeutic applications.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Hutchins
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bessie Meechoovet
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ritin Sharma
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Natacha Carnel-Amar
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claire Lin
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher Azzam
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ima Ghaeli
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Kaushik V Amancherla
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - José Fabian Victorino
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Krystine Garcia-Mansfield
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ryan Pfeffer
- Masonic Medical Research Institute, Utica, NY, USA 13501
| | - Parul Sahu
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian R. Lindman
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sammy Elmariah
- Department of Medicine, Cardiovascular Division, University of California-San Francisco, San Francisco, CA, USA
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Michael J. Betti
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Xavier Bledsoe
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | | | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yan Ru Su
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ngoc Do
- Spectradyne LLC, Signal Hill, CA, USA
| | - Marta Garcia Contreras
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dimitrios Varrias
- Albert Einstein College of Medicine/ Jacobi Medical Center, The Bronx, NY, USA
| | - Michail Kladas
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Miroslav Radulovic
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Dimitris Tsiachris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | | | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - James L. Januzzi
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Ravi Shah
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Webber K, Patel S, Kizer JR, Eastell R, Psaty BM, Newman AB, Cummings SR. Associations of Serum GDF-15 Levels with Physical Performance, Mobility Disability, Cognition, Cardiovascular Disease, and Mortality in Older Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311629. [PMID: 39148825 PMCID: PMC11326340 DOI: 10.1101/2024.08.07.24311629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Growth differentiation factor 15 (GDF-15) is a member of the TGFβ superfamily secreted by many cell types and found at higher blood concentrations as chronological age increases (1). Given the emergence of GDF-15 as a key protein associated with aging, it is important to understand the multitude of conditions with which circulating GDF-15 is associated. Methods We pooled data from 1,174 randomly selected Health ABC Study (Health ABC) participants and 1,503 Cardiovascular Health Study (CHS) participants to evaluate the risk of various conditions and age-related outcomes across levels of GDF-15. The primary outcomes were (1) risk of mobility disability and falls; (2) impaired cognitive function; (3) and increased risk of cardiovascular disease and total mortality. Results The pooled study cohort had a mean age of 75.4 +/-4.4 years. Using a Bonferroni-corrected threshold, our analyses show that high levels of GDF-15 were associated with a higher risk of severe mobility disability (HR: 2.13 [1.64, 2.77]), coronary heart disease (HR: 1.47 [1.17, 1.83]), atherosclerotic cardiovascular disease (HR: 1.56 [1.22, 1.98]), heart failure (HR: 2.09 [1.66, 2.64]), and mortality (HR: 1.81 [1.53, 2.15]) when comparing the highest and lowest quartiles. For CHS participants, analysis of extreme quartiles in fully adjusted models revealed a 3.5-fold higher risk of dementia (HR: 3.50 [1.97, 6.22]). Conclusions GDF-15 is associated with several age-related outcomes and diseases, including mobility disability, impaired physical and cognitive performance, dementia, cardiovascular disease, and mortality. Each of these findings demonstrates the importance of GDF-15 as a potential biomarker for many aging-related conditions.
Collapse
Affiliation(s)
- Katey Webber
- Research Institute, California Pacific Medical Center, San Francisco, CA
- Institute on Aging, San Francisco, CA
| | - Sheena Patel
- Research Institute, California Pacific Medical Center, San Francisco, CA
| | - Jorge R. Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Department of Medicine, University of California, San Francisco, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Richard Eastell
- Department of Oncology and Metabolism, Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Heath Systems and Population Health, University of Washington, Seattle, WA
| | - Anne B. Newman
- Center for Aging and Population Health, Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Steven R. Cummings
- Research Institute, California Pacific Medical Center, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
17
|
Lee MM, Zuo Y, Steiling K, Mizgerd JP, Kalesan B, Walkey AJ. Clinical risk factors and blood protein biomarkers of 10-year pneumonia risk. PLoS One 2024; 19:e0296139. [PMID: 38968193 PMCID: PMC11226120 DOI: 10.1371/journal.pone.0296139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/26/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Chronic inflammation may increase susceptibility to pneumonia. RESEARCH QUESTION To explore associations between clinical comorbidities, serum protein immunoassays, and long-term pneumonia risk. METHODS Framingham Heart Study Offspring Cohort participants ≥65 years were linked to their Centers for Medicare Services claims data. Clinical data and 88 serum protein immunoassays were evaluated for associations with 10-year incident pneumonia risk using Fine-Gray models for competing risks of death and least absolute shrinkage and selection operators for covariate selection. RESULTS We identified 1,370 participants with immunoassays and linkage to Medicare data. During 10 years of follow up, 428 (31%) participants had a pneumonia diagnosis. Chronic pulmonary disease [subdistribution hazard ratio (SHR) 1.87; 95% confidence interval (CI), 1.33-2.61], current smoking (SHR 1.79, CI 1.31-2.45), heart failure (SHR 1.74, CI 1.10-2.74), atrial fibrillation/flutter (SHR 1.43, CI 1.06-1.93), diabetes (SHR 1.36, CI 1.05-1.75), hospitalization within one year (SHR 1.34, CI 1.09-1.65), and age (SHR 1.06 per year, CI 1.04-1.08) were associated with pneumonia. Three baseline serum protein measurements were associated with pneumonia risk independent of measured clinical factors: growth differentiation factor 15 (SHR 1.32; CI 1.02-1.69), C-reactive protein (SHR 1.16, CI 1.06-1.27) and matrix metallopeptidase 8 (SHR 1.14, CI 1.01-1.30). Addition of C-reactive protein to the clinical model improved prediction (Akaike information criterion 4950 from 4960; C-statistic of 0.64 from 0.62). CONCLUSIONS Clinical comorbidities and serum immunoassays were predictive of pneumonia risk. C-reactive protein, a routinely-available measure of inflammation, modestly improved pneumonia risk prediction over clinical factors. Our findings support the hypothesis that prior inflammation may increase the risk of pneumonia.
Collapse
Affiliation(s)
- Ming-Ming Lee
- Pulmonary and Critical Care Medicine, Norwalk Hospital, Nuvance Health, Norwalk, CT, United States of America
| | - Yi Zuo
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States of America
| | - Katrina Steiling
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Joseph P. Mizgerd
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Bindu Kalesan
- Boston University School of Medicine, Boston, MA, United States of America
| | - Allan J. Walkey
- Division of Health Systems Science, Department of Medicine, UMass Chan Medical School, Worcester, MA, United States of America
| |
Collapse
|
18
|
Zhang YS, Liu ZY, Liu ZY, Lin LC, Chen Q, Zhao JY, Tao H. m6A epitranscriptomic modification of inflammation in cardiovascular disease. Int Immunopharmacol 2024; 134:112222. [PMID: 38728881 DOI: 10.1016/j.intimp.2024.112222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 04/28/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Cardiovascular disease is currently the number one cause of death endangering human health. There is currently a large body of research showing that the development of cardiovascular disease and its complications is often accompanied by inflammatory processes. In recent years, epitranscriptional modifications have been shown to be involved in regulating the pathophysiological development of inflammation in cardiovascular diseases, with 6-methyladenine being one of the most common RNA transcriptional modifications. In this review, we link different cardiovascular diseases, including atherosclerosis, heart failure, myocardial infarction, and myocardial ischemia-reperfusion, with inflammation and describe the regulatory processes involved in RNA methylation. Advances in RNA methylation research have revealed the close relationship between the regulation of transcriptome modifications and inflammation in cardiovascular diseases and brought potential therapeutic targets for disease diagnosis and treatment. At the same time, we also discussed different cell aspects. In addition, in the article we also describe the different application aspects and clinical pathways of RNA methylation therapy. In summary, this article reviews the mechanism, regulation and disease treatment effects of m6A modification on inflammation and inflammatory cells in cardiovascular diseases in recent years. We will discuss issues facing the field and new opportunities that may be the focus of future research.
Collapse
Affiliation(s)
- Yun-Sen Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Qi Chen
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
19
|
Huang X, Mu M, Wang B, Zhang H, Liu Y, Yu L, Zhou M, Ma J, Wang D, Chen W. Associations of coal mine dust exposure with arterial stiffness and atherosclerotic cardiovascular disease risk in chinese coal miners. Int Arch Occup Environ Health 2024; 97:473-484. [PMID: 38530481 DOI: 10.1007/s00420-024-02062-2] [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: 10/18/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE Whether coal mine dust exposure increases cardiovascular diseases (CVDs) risk was rarely explored. Our objective was to examine the association between coal mine dust exposure and cardiovascular risk. METHODS We estimated cumulative coal mine dust exposure (CDE) for 1327 coal miners by combining data on workplace dust concentrations and work history. We used brachial-ankle pulse wave velocity (baPWV, a representative indicator of arterial stiffness) and ten-year atherosclerotic cardiovascular disease (ASCVD) risk to assess potential CVD risk, exploring their associations with CDE. RESULTS Positive dose-response relationships of CDE with baPWV and ten-year ASCVD risk were observed after adjusting for covariates. Specifically, each 1 standard deviation (SD) increase in CDE was related to a 0.27 m/s (95% CI: 0.21, 0.34) increase in baPWV and a 1.29 (95% CI: 1.14, 1.46) elevation in OR (odds ratio) of risk of abnormal baPWV. Moreover, each 1 SD increase in CDE was associated with a 0.74% (95% CI: 0.63%, 0.85%) increase in scores of ten-year ASCVD and a 1.91 (95% CI: 1.62, 2.26) increase in OR of risk of ten-year ASCVD. When compared with groups unexposed to coal mine dust, significant increase in the risk of arterial stiffness and ten-year ASCVD in the highest CDE groups were detected. CONCLUSION The study suggested that cumulative exposure to coal mine dust was associated with elevated arterial stiffness and ten-year ASCVD risk in a dose-response manner. These findings contribute valuable insights for cardiovascular risk associated with coal mine dust.
Collapse
Affiliation(s)
- Xuezan Huang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, 232000, Anhui, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Haozhe Zhang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yang Liu
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Linling Yu
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Min Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongming Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
20
|
Robbertse PPS, Steyn J, Rajah MR, Doubell AF, Nachega JB, Herbst PG. Subclinical cardiovascular remodelling in HIV-infection: A multimodal case study of 2 serodiscordant, monozygotic twins. SA HEART JOURNAL 2024; 21:48-57. [PMID: 38737401 PMCID: PMC11087033 DOI: 10.24170/21-1-6322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Cardiovascular abnormalities are increasingly recognised among people newly diagnosed with HIV, but subclinical pathology may be challenging to diagnose. We present a case study of subtle cardiovascular changes in identical twins, one without HIV-infection and the other recently diagnosed with HIV (serodiscordant). We hypothesise that cardiovascular parameters would be similar between the twins, unless non-genetic (environmental) factors are at play. These differences likely represent occult pathology secondary to the effects of early HIV-infection. A 25-year-old female incidentally diagnosed with HIV, and her HIV-uninfected identical twin, living with her since birth, underwent comprehensive cardiovascular assessments. The HIV-positive twin exhibited a globular left ventricle (LV), larger LV volumes, decreased LV strain, peak atrial longitudinal strain (PALS) and higher native T1 and T2 mapping values compared to her sister. Cardiac biomarkers high sensitivity cardiac troponin T and N-terminal proBNP, as well as the novel markers of fibrosis and remodelling, galectin-3 and soluble-ST2, were higher in the HIV-infected twin. Given the twins' shared environment and genetic makeup, these differences likely stem from HIV-infection. Our study supports previous findings and suggests potential screening markers for HIV-associated cardiovascular disease, including PALS. Further research is warranted to explore PALS' utility in this context.
Collapse
Affiliation(s)
- Pieter-Paul S. Robbertse
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, South Africa
- University of Pittsburgh HIV-Comorbidities Research Training Programme in South Africa
| | - Jan Steyn
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, South Africa
| | - Megan R. Rajah
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, South Africa
| | - Anton F. Doubell
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, South Africa
| | - Jean B. Nachega
- University of Pittsburgh HIV-Comorbidities Research Training Programme in South Africa
- Department of Medicine and Centre for Infectious Diseases, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
- Department of Epidemiology and International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Epidemiology, Infectious Diseases and Microbiology, and Centre for Global Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Philip G. Herbst
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, South Africa
| |
Collapse
|
21
|
Chen YJ, Chou CY, Er TK. Correlations of sST2 and Gal-3 with Cardiothoracic Ratio in Patients with Chronic Kidney Disease. Biomedicines 2024; 12:791. [PMID: 38672149 PMCID: PMC11048335 DOI: 10.3390/biomedicines12040791] [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: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic kidney disease (CKD) frequently correlates with cardiovascular complications. Soluble suppression of tumorigenicity 2 (sST2) and Galectin-3 (Gal-3) are emerging as cardiac markers with potential relevance in cardiovascular risk prediction. The cardiothoracic ratio (CTR), a metric easily obtainable from chest radiographs, has traditionally been used to assess cardiac size and the potential for cardiomegaly. Understanding the correlation between these cardiac markers and the cardiothoracic ratio (CTR) could provide valuable insights into the cardiovascular prognosis of CKD patients. This study aimed to explore the relationship between sST2, Gal-3, and the CTR in individuals with CKD. Plasma concentrations of sST2 and Gal-3 were assessed in a cohort of 123 CKD patients by enzyme-linked immunosorbent assay (ELISA). On a posterior-to-anterior chest X-ray view, the CTR was determined by comparing the widths of the heart to that of the thorax. The mean concentration of sST2 in the study participants ranged from 775.4 to 4475.6 pg/mL, and the mean concentration of Gal-3 ranged from 4.7 to 9796.0 ng/mL. Significant positive correlations were observed between sST2 and the CTR (r = 0.291, p < 0.001) and between Gal-3 and the CTR (r = 0.230, p < 0.01). Our findings indicate that elevated levels of sST2 and Gal-3 are associated with an increased CTR in CKD patients. This relationship may enable better cardiovascular risk evaluation for CKD patients. Further studies are warranted to explore the clinical implications of these associations.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung 41354, Taiwan
| | - Che-Yi Chou
- Division of Nephrology, Asia University, Taichung 41354, Taiwan
| | - Tze-Kiong Er
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung 41354, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
22
|
Robbertse PS, Doubell AF, Esterhuizen TM, Herbst PG. Antiretroviral therapy and HIV-associated cardiovascular disease: a prospective cardiac biomarker and CMR tissue characterization study. ESC Heart Fail 2024; 11:748-758. [PMID: 38100145 PMCID: PMC10966217 DOI: 10.1002/ehf2.14603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 08/01/2023] [Accepted: 11/10/2023] [Indexed: 02/24/2024] Open
Abstract
AIMS Biochemical markers are fundamental in cardiac evaluation, and various novel assays have recently been discovered. We prospectively evaluated the hearts of newly diagnosed people living with human immunodeficiency virus (PLWH) using cardiac biomarkers, compared them with human immunodeficiency virus (HIV)-uninfected controls, and correlated our prospective findings with cardiovascular magnetic resonance imaging (CMR). METHODS AND RESULTS Newly diagnosed, antiretroviral therapy (ART)-naïve PLWH were recruited along with HIV-uninfected, age-matched, and sex-matched controls. All participants underwent measurement of high-sensitivity cardiac troponin T (hs-cTnT), N-terminal pro-B-type natriuretic peptide (NT-proBNP), soluble ST2 (sST2), and galectin-3, as well as a CMR study with multiparametric mapping. The HIV group started ART and was re-evaluated 9 months later. The cardiac biomarkers and their correlation with CMR parameters were evaluated in and between groups. Compared with controls (n = 22), hs-cTnT (4.0 vs. 5.1 ng/L; P = 0.004), NT-proBNP (23.2 vs. 40.8 ng/L; P = 0.02), and galectin-3 (6.8 vs. 9.0 ng/mL; P = 0.002) were all significantly higher in the ART-naïve group (n = 73). After 9 months of ART, hs-cTnT (5.1 vs. 4.3 ng/L; P = 0.02) and NT-proBNP (40.8 vs. 28.5 ng/L; P = 0.03) both decreased significantly and a trend of decrease was seen in sST2 (16.5 vs. 14.8 ng/L; P = 0.08). Galectin-3 did not demonstrate decrease over time (9.0 vs. 8.8 ng/mL; P = 0.6). The cardiac biomarkers that showed the best correlation with CMR measurements native T1, T2, and extracellular volume were NT-proBNP (rs ≥ 0.4, P < 0.001) and galectin-3 (rs ≥ 0.3, P < 0.01). CONCLUSIONS Our cardiac biomarker data support the presence of subclinical myocardial injury, remodelling, and fibrosis at HIV diagnosis, and ART had a positive influence on these blood markers. It remains unclear if the underlying pathological processes were fully addressed by ART. The ability of cardiac biomarkers to detect and track tissue abnormalities diagnosed with CMR showed promise. With additional research, this could lead to improvements in screening and monitoring myocardial abnormalities, even in CMR-limited settings.
Collapse
Affiliation(s)
- Pieter‐Paul S. Robbertse
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch University and Tygerberg HospitalCape TownSouth Africa
- University of Pittsburgh HIV‐Comorbidities Research Training Programme in South AfricaCape TownSouth Africa
| | - Anton F. Doubell
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch University and Tygerberg HospitalCape TownSouth Africa
| | - Tonya M. Esterhuizen
- Division of Epidemiology and Biostatistics, Department of Global HealthStellenbosch UniversityStellenboschSouth Africa
| | - Philip G. Herbst
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch University and Tygerberg HospitalCape TownSouth Africa
| |
Collapse
|
23
|
Yang P, Wang S, Zhong C, Yin J, Yang J, Wang A, Xu T, Zhang Y. Association of Cardiac Biomarkers in Combination With Cognitive Impairment After Acute Ischemic Stroke. J Am Heart Assoc 2024; 13:e031010. [PMID: 38390800 PMCID: PMC10944059 DOI: 10.1161/jaha.123.031010] [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: 05/16/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Poststroke cognitive impairment is a severe and common clinical complication that constitutes a substantial global health burden. We aimed to evaluate the association of 3 cardiac biomarkers in combination with poststroke cognitive impairment and their prognostic significance. METHODS AND RESULTS This prospective study included 566 patients with ischemic stroke. Cardiac biomarkers, including sST2 (soluble suppression of tumorigenicity-2 receptor), GDF-15 (growth differentiation factor-15), and NT-proBNP (N-terminal pro-B-type natriuretic peptide), were measured. Cognitive impairment was defined as a Mini-Mental State Examination score of <27 or a Montreal Cognitive Assessment score of <25 at 3 months after ischemic stroke. Odds of cognitive impairment 3 months after ischemic stroke increased with the number of elevated cardiac biomarkers (sST2, GDF-15, and NT-proBNP; Ptrend<0.001). The multivariable adjusted odds ratios (95% CIs) of cognitive impairment defined by the Mini-Mental State Examination and Montreal Cognitive Assessment were 2.45 (1.48-4.07) and 1.86 (1.10-3.14) for the participants with ≥2 elevated cardiac biomarkers, respectively, compared with those without any elevated cardiac biomarker. Additionally, higher cardiac biomarker scores were associated with an increased risk of cognitive impairment (Ptrend<0.05). Simultaneously adding all 3 cardiac biomarkers to the basic model with traditional risk factors significantly improved the risk prediction of Mini-Mental State Examination-defined cognitive impairment (net reclassification improvement=34.99%, P<0.001; integrated discrimination index=2.67%, P<0.001). Similar findings were observed using the Montreal Cognitive Assessment scores. CONCLUSIONS An increased number of elevated novel cardiac biomarkers were associated with an increased odds of poststroke cognitive impairment, suggesting that a combination of these cardiac biomarkers may improve the risk prediction of cognitive impairment. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT01840072.
Collapse
Affiliation(s)
- Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Shuyao Wang
- Department of NeurologyTongliao Municipal HospitalTongliaoChina
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Jieyun Yin
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Jingyuan Yang
- Department of Epidemiology, School of Public HealthGuizhou Medical UniversityGuiyangChina
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSuzhou Medical College of Soochow UniversitySuzhouChina
| |
Collapse
|
24
|
Welsh P, Kimenai DM, Shah ASV, Gadd DA, Marioni RE, Woodward M, Sudlow CLM, Campbell A, Cleland JGF, Pellicori P, Hayward C, Mills NL, Sattar N. Multiple Cardiac Biomarkers to Improve Prediction of Cardiovascular Events: Findings from the Generation Scotland Scottish Family Health Study. Clin Chem 2024; 70:403-413. [PMID: 38069915 DOI: 10.1093/clinchem/hvad205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Many studies have investigated whether single cardiac biomarkers improve cardiovascular risk prediction for primary prevention but whether a combined approach could further improve risk prediction is unclear. We aimed to test a sex-specific, combined cardiac biomarker approach for cardiovascular risk prediction. METHODS In the Generation Scotland Scottish Family Health Study, N-terminal pro-B-type natriuretic peptide (NT-proBNP), growth differentiation factor-15 (GDF-15), cardiac troponin I (cTnI), cardiac troponin T (cTnT), and C-reactive protein (CRP) were measured in stored serum using automated immunoassays. Sex-specific Cox models that included SCORE2 risk factors evaluated addition of single and combined biomarkers for prediction of major adverse cardiovascular events (MACE). Combined biomarker models were compared to a baseline model that included SCORE2 risk factors. RESULTS The study population comprised 18 383 individuals (58.9% women, median age of 48 years [25th-75th percentile, 35-58 years]). During the median follow up of 11.6 (25th-75th percentile, 10.8-13.0) years, MACE occurred in 942 (5.1%) individuals. The greatest increase in discrimination with addition of individual biomarkers to the base model was for women GDF-15 and for men NT-proBNP (change in c-index: + 0.010 for women and +0.005 for men). For women, combined biomarker models that included GDF-15 and NT-proBNP (+0.012) or GDF-15 and cTnI (+0.013), but not CRP or cTnT, further improved discrimination. For men, combined biomarker models that included NT-proBNP and GDF-15 (+0.007), NT-proBNP and cTnI (+0.006), or NT-proBNP and CRP (+0.008), but not cTnT, further improved discrimination. CONCLUSIONS A combined biomarker approach, particularly the use of GDF-15, NT-proBNP and cTnI, further refined cardiovascular risk estimates.
Collapse
Affiliation(s)
- Paul Welsh
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Dorien M Kimenai
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Anoop S V Shah
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Danni A Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Woodward
- The George Institute for Global Health, School of Public Health, Imperial College London, London, United Kingdom
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Cathie L M Sudlow
- British Heart Foundation Data Science Centre, Health Data Research UK, London, United Kingdom
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - John G F Cleland
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Pierpaolo Pellicori
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Naveed Sattar
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
25
|
Kayani M, Fatima N, Yarra PC, Almansouri NE, K D, Balasubramanian A, Parvathaneni N, Mowo-Wale AG, Valdez JA, Nazir Z. Novel Biomarkers in Early Detection of Heart Failure: A Narrative Review. Cureus 2024; 16:e53445. [PMID: 38435138 PMCID: PMC10909379 DOI: 10.7759/cureus.53445] [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] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Heart failure (HF) represents a significant global health challenge, characterized by a variety of symptoms resulting from cardiac dysfunction. This dysfunction often leads to systemic and pulmonary congestion. The pathophysiology of HF is complex, involving stimulation of the sympathetic nervous system, which is insufficiently balanced by the release of natriuretic peptide. This imbalance leads to progressive hypertrophy and dilatation of the heart's chambers, impairing its pumping efficiency and increasing the risk of arrhythmias and conduction disorders. The prevalence of HF is exceptionally high in industrialized nations and is expected to increase owing to an aging population and advancements in diagnostic methods. This study emphasizes the critical role of early diagnosis in reducing morbidity and mortality associated with HF, focusing specifically on the evolving importance of biomarkers in managing this condition. Biomarkers have played a key role in transforming the diagnosis and treatment of HF. Traditional biomarkers such as b-type natriuretic peptide and N-terminal pro-b-type natriuretic peptide have been widely adopted for their cost-effectiveness and ease of access. However, the rise of novel biomarkers such as growth differentiation factor 15 and adrenomedullin has shown promising results, offering superior sensitivity and specificity. These new biomarkers enhance diagnostic accuracy, risk stratification, and prognostic evaluation in HF patients. Despite these advancements, challenges remain, such as limited availability, high costs, and the need for further validation in diverse patient populations. Through a comprehensive literature review across databases such as PubMed, Google Scholar, and the Cochrane Library, this study compiles and analyzes data from 18 relevant studies, offering a detailed understanding of the current state of HF biomarkers. The study examines both traditional and emerging biomarkers such as galectin-3 and soluble suppression of tumorigenicity 2 in HF, exploring their clinical roles and impact on patient outcomes.
Collapse
Affiliation(s)
- Maryam Kayani
- Cardiology, Shifa Tameer-e-Millat University Shifa College of Medicine, Islamabad, PAK
| | - Neha Fatima
- Internal Medicine, Lisie Hospital, Kochi, IND
| | | | - Naiela E Almansouri
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, University of Tripoli, Tripoli, LBY
| | - Deepshikha K
- Cardiology, Pondicherry Institute of Medical Sciences, Pondicherry, IND
| | | | | | | | - Josue A Valdez
- General Practice, Universidad Autónoma de Durango, Los Mochis, MEX
| | - Zahra Nazir
- Internal Medicine, Combined Military Hospital, Quetta, PAK
| |
Collapse
|
26
|
Sebastian SA, Co EL, Mahtani A, Padda I, Anam M, Mathew SS, Shahzadi A, Niazi M, Pawar S, Johal G. Heart Failure: Recent Advances and Breakthroughs. Dis Mon 2024; 70:101634. [PMID: 37704531 DOI: 10.1016/j.disamonth.2023.101634] [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] [Indexed: 09/15/2023]
Abstract
Heart failure (HF) is a common clinical condition encountered in various healthcare settings with a vast socioeconomic impact. Recent advancements in pharmacotherapy have led to the evolution of novel therapeutic agents with a decrease in hospitalization and mortality rates in HF with reduced left ventricular ejection fraction (HFrEF). Lately, the introduction of artificial intelligence (AI) to construct decision-making models for the early detection of HF has played a vital role in optimizing cardiovascular disease outcomes. In this review, we examine the newer therapies and evidence behind goal-directed medical therapy (GDMT) for managing HF. We also explore the application of AI and machine learning (ML) in HF, including early diagnosis and risk stratification for HFrEF.
Collapse
Affiliation(s)
| | - Edzel Lorraine Co
- University of Santo Tomas Faculty of Medicine and Surgery, Manila, Philippines
| | - Arun Mahtani
- Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Inderbir Padda
- Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Mahvish Anam
- Deccan College of Medical Sciences, Hyderabad, India
| | | | | | - Maha Niazi
- Royal Alexandra Hospital, Edmonton, Canada
| | | | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, Washington, USA
| |
Collapse
|
27
|
Linders AN, Dias IB, López Fernández T, Tocchetti CG, Bomer N, Van der Meer P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. NPJ AGING 2024; 10:9. [PMID: 38263284 PMCID: PMC10806194 DOI: 10.1038/s41514-024-00135-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
The population of cancer survivors is rapidly increasing due to improving healthcare. However, cancer therapies often have long-term side effects. One example is cancer therapy-related cardiac dysfunction (CTRCD) caused by doxorubicin: up to 9% of the cancer patients treated with this drug develop heart failure at a later stage. In recent years, doxorubicin-induced cardiotoxicity has been associated with an accelerated aging phenotype and cellular senescence in the heart. In this review we explain the evidence of an accelerated aging phenotype in the doxorubicin-treated heart by comparing it to healthy aged hearts, and shed light on treatment strategies that are proposed in pre-clinical settings. We will discuss the accelerated aging phenotype and the impact it could have in the clinic and future research.
Collapse
Affiliation(s)
- Annet Nicole Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Itamar Braga Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Teresa López Fernández
- Division of Cardiology, Cardiac Imaging and Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Federico II University, Naples, Italy
- Centre for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
- Interdepartmental Centre of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Centre (CIRIAPA), Federico II University, Naples, Italy
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands.
| |
Collapse
|
28
|
Patel KV, Segar MW, Klonoff DC, Khan MS, Usman MS, Lam CSP, Verma S, DeFilippis AP, Nasir K, Bakker SJL, Westenbrink BD, Dullaart RPF, Butler J, Vaduganathan M, Pandey A. Optimal Screening for Predicting and Preventing the Risk of Heart Failure Among Adults With Diabetes Without Atherosclerotic Cardiovascular Disease: A Pooled Cohort Analysis. Circulation 2024; 149:293-304. [PMID: 37950893 PMCID: PMC11257100 DOI: 10.1161/circulationaha.123.067530] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/01/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND The optimal approach to identify individuals with diabetes who are at a high risk for developing heart failure (HF) to inform implementation of preventive therapies is unknown, especially in those without atherosclerotic cardiovascular disease (ASCVD). METHODS Adults with diabetes and no HF at baseline from 7 community-based cohorts were included. Participants without ASCVD who were at high risk for developing HF were identified using 1-step screening strategies: risk score (WATCH-DM [Weight, Age, Hypertension, Creatinine, HDL-C, Diabetes Control, QRS Duration, MI, and CABG] ≥12), NT-proBNP (N-terminal pro-B-type natriuretic peptide ≥125 pg/mL), hs-cTn (high-sensitivity cardiac troponin T ≥14 ng/L; hs-cTnI ≥31 ng/L), and echocardiography-based diabetic cardiomyopathy (echo-DbCM; left atrial enlargement, left ventricular hypertrophy, or diastolic dysfunction). High-risk participants were also identified using 2-step screening strategies with a second test to identify residual risk among those deemed low risk by the first test: WATCH-DM/NT-proBNP, NT-proBNP/hs-cTn, NT-proBNP/echo-DbCM. Across screening strategies, the proportion of HF events identified, 5-year number needed to treat and number needed to screen to prevent 1 HF event with an SGLT2i (sodium-glucose cotransporter 2 inhibitor) among high-risk participants, and cost of screening were estimated. RESULTS The initial study cohort included 6293 participants (48.2% women), of whom 77.7% without prevalent ASCVD were evaluated with different HF screening strategies. At 5-year follow-up, 6.2% of participants without ASCVD developed incident HF. The 5-year number needed to treat to prevent 1 HF event with an SGLT2i among participants without ASCVD was 43 (95% CI, 29-72). In the cohort without ASCVD, high-risk participants identified using 1-step screening strategies had a low 5-year number needed to treat (22 for NT-proBNP to 37 for echo-DbCM). However, a substantial proportion of HF events occurred among participants identified as low risk using 1-step screening approaches (29% for echo-DbCM to 47% for hs-cTn). Two-step screening strategies captured most HF events (75-89%) in the high-risk subgroup with a comparable 5-year number needed to treat as the 1-step screening approaches (30-32). The 5-year number needed to screen to prevent 1 HF event was similar across 2-step screening strategies (45-61). However, the number of tests and associated costs were lowest for WATCH-DM/NT-proBNP ($1061) compared with other 2-step screening strategies (NT-proBNP/hs-cTn: $2894; NT-proBNP/echo-DbCM: $16 358). CONCLUSIONS Selective NT-proBNP testing based on the WATCH-DM score efficiently identified a high-risk primary prevention population with diabetes expected to derive marked absolute benefits from SGLT2i to prevent HF.
Collapse
Affiliation(s)
- Kershaw V. Patel
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Matthew W. Segar
- Department of Cardiology, Texas Heart Institute, Houston, TX, USA
| | - David C. Klonoff
- Diabetes Research Institute, Mills-Peninsula Medical Center, San Mateo, CA, USA
| | - Muhammad Shahzeb Khan
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Muhammad Shariq Usman
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carolyn S. P. Lam
- National Heart Centre Singapore, Duke-National University of Singapore, Singapore
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Andrew P. DeFilippis
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Khurram Nasir
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Stephan J. L. Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, The Netherlands
| | - B. Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robin P. F. Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Baylor Scott and White Research Institute, Dallas, Texas, USA
| | - Muthiah Vaduganathan
- Brigham and Women’s Hospital Heart and Vascular Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
29
|
Shah AM, Myhre PL, Arthur V, Dorbala P, Rasheed H, Buckley LF, Claggett B, Liu G, Ma J, Nguyen NQ, Matsushita K, Ndumele C, Tin A, Hveem K, Jonasson C, Dalen H, Boerwinkle E, Hoogeveen RC, Ballantyne C, Coresh J, Omland T, Yu B. Large scale plasma proteomics identifies novel proteins and protein networks associated with heart failure development. Nat Commun 2024; 15:528. [PMID: 38225249 PMCID: PMC10789789 DOI: 10.1038/s41467-023-44680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Heart failure (HF) causes substantial morbidity and mortality but its pathobiology is incompletely understood. The proteome is a promising intermediate phenotype for discovery of novel mechanisms. We measured 4877 plasma proteins in 13,900 HF-free individuals across three analysis sets with diverse age, geography, and HF ascertainment to identify circulating proteins and protein networks associated with HF development. Parallel analyses in Atherosclerosis Risk in Communities study participants in mid-life and late-life and in Trøndelag Health Study participants identified 37 proteins consistently associated with incident HF independent of traditional risk factors. Mendelian randomization supported causal effects of 10 on HF, HF risk factors, or left ventricular size and function, including matricellular (e.g. SPON1, MFAP4), senescence-associated (FSTL3, IGFBP7), and inflammatory (SVEP1, CCL15, ITIH3) proteins. Protein co-regulation network analyses identified 5 modules associated with HF risk, two of which were influenced by genetic variants that implicated trans hotspots within the VTN and CFH genes.
Collapse
Affiliation(s)
- Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peder L Myhre
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pranav Dorbala
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Humaira Rasheed
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Guning Liu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jianzhong Ma
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Adrienne Tin
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristian Hveem
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian Jonasson
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Cardiology, St Olavs University Hospital, Trondheim, Norway
- Department of Internal Medicine, Levanger Hospital, Levanger, Norway
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ron C Hoogeveen
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Josef Coresh
- Departments of Medicine and Population Health, NYU Langone Health, New York, NY, USA
| | - Torbjørn Omland
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| |
Collapse
|
30
|
Balaguer J, García-Foncillas J, Tuñón J. Natriuretic peptides: Another tool for the management of cancer? Crit Rev Oncol Hematol 2024; 193:104219. [PMID: 38029944 DOI: 10.1016/j.critrevonc.2023.104219] [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: 07/08/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023] Open
Abstract
The connection between heart failure (HF) and cancer through multiple pathways such as inflammation, oxidative stress, and neurohormonal activation, among others, is well established. As a consequence, increases in plasma levels of several biomarkers have been described in both disorders. The most consistent information is related to natriuretic peptides (NPs). Although they are known to be produced in the ventricles as a response to myocardial distension, and thus can be useful for the diagnosis and prognosis of HF, and also for the management of chemotherapy-induced myocardial damage, they are also produced by tumour cells. In this regard, increased plasma levels of NPs have been described in patients with multiple malignancies in the absence of volume overload. Natriuretic peptide levels have been shown to correlate directly with the extension of tumours and with poorer outcomes. Moreover, some data indicate that they may help in the detection of subclinical tumours. Given that these peptides have been described to have anti-proliferative and anti-angiogenic effects, a plausible hypothesis is that they may be produced by tumours as a negative feed-back mechanism to avoid tumour progression. This would lead to increased levels of NPs in plasma that could be potentially useful for early detection of malignancies as well as for a prognostic assessment. Nevertheless, since the sample size of many studies published so far is limited, more data are needed to provide consistent data in order to confirm or rule out this hypothesis.
Collapse
Affiliation(s)
- Jorge Balaguer
- Division of Cardiology, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Jesús García-Foncillas
- Division of Oncology, IIS-Fundación Jiménez Díaz, Madrid, Spain; School of Medicine, Autónoma University, Madrid, Spain
| | - José Tuñón
- Division of Cardiology, IIS-Fundación Jiménez Díaz, Madrid, Spain; School of Medicine, Autónoma University, Madrid, Spain; Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, Madrid, Spain; CIBERCV, ISCIII, Madrid, Spain.
| |
Collapse
|
31
|
Lee MM, Zuo Y, Steiling K, Mizgerd JP, Kalesan B, Walkey AJ. Clinical risk factors and blood protein biomarkers of 10-year pneumonia risk. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.07.23299678. [PMID: 38105941 PMCID: PMC10723561 DOI: 10.1101/2023.12.07.23299678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Chronic inflammation may increase susceptibility to pneumonia. Research Question To explore associations between clinical comorbidities, serum protein immunoassays, and long-term pneumonia risk. Methods Framingham Heart Study Offspring Cohort participants ≥65 years were linked to their Centers for Medicare Services claims data. Clinical data and 88 serum protein immunoassays were evaluated for associations with 10-year incident pneumonia risk using Fine-Gray models for competing risks of death and least absolute shrinkage and selection operators for covariate selection. Results We identified 1,370 participants with immunoassays and linkage to Medicare data. During 10 years of follow up, 428 (31%) participants had a pneumonia diagnosis. Chronic pulmonary disease [subdistribution hazard ratio (SHR) 1.87; 95% confidence interval (CI), 1.33-2.61], current smoking (SHR 1.79, CI 1.31-2.45), heart failure (SHR 1.74, CI 1.10-2.74), atrial fibrillation/flutter (SHR 1.43, CI 1.06-1.93), diabetes (SHR 1.36, CI 1.05-1.75), hospitalization within one year (SHR 1.34, CI 1.09-1.65), and age (SHR 1.06 per year, CI 1.04-1.08) were associated with pneumonia. Three baseline serum protein measurements were associated with pneumonia risk independent of measured clinical factors: growth differentiation factor 15 (SHR 1.32; CI 1.02-1.69), C-reactive protein (SHR 1.16, CI 1.06-1.27) and matrix metallopeptidase 8 (SHR 1.14, CI 1.01-1.30). Addition of C-reactive protein to the clinical model improved prediction (Akaike information criterion 4950 from 4960; C-statistic of 0.64 from 0.62). Conclusions Clinical comorbidities and serum immunoassays were predictive of pneumonia risk. C-reactive protein, a routinely-available measure of inflammation, modestly improved pneumonia risk prediction over clinical factors. Our findings support the hypothesis that prior inflammation may increase the risk of pneumonia.
Collapse
Affiliation(s)
- Ming-Ming Lee
- Pulmonary and Critical Care Medicine, Norwalk Hospital, Nuvance Health, Norwalk, CT
| | - Yi Zuo
- Department of Biostatistics, Vanderbilt University, Nashville, TN
| | - Katrina Steiling
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston MA
| | - Joseph P. Mizgerd
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA
| | | | - Allan J. Walkey
- Division of Health Systems Science, Department of Medicine, UMass Chan Medical School, Worcester, MA
| |
Collapse
|
32
|
Kotta PA, Nambi V, Bozkurt B. Biomarkers for Heart Failure Prediction and Prevention. J Cardiovasc Dev Dis 2023; 10:488. [PMID: 38132656 PMCID: PMC10744096 DOI: 10.3390/jcdd10120488] [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: 09/17/2023] [Revised: 11/13/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Heart failure (HF) is a global pandemic affecting over 64 million people worldwide. Its prevalence is on an upward trajectory, with associated increasing healthcare expenditure. Organizations including the American College of Cardiology (ACC) and the American Heart Association (AHA) have identified HF prevention as an important focus. Recently, the ACC/AHA/Heart Failure Society of America (HFSA) Guidelines on heart failure were updated with a new Class IIa, Level of Evidence B recommendation for biomarker-based screening in patients at risk of developing heart failure. In this review, we evaluate the studies that have assessed the various roles and contributions of biomarkers in the prediction and prevention of heart failure. We examined studies that have utilized biomarkers to detect cardiac dysfunction or abnormality for HF risk prediction and screening before patients develop clinical signs and symptoms of HF. We also included studies with biomarkers on prognostication and risk prediction over and above existing HF risk prediction models and studies that address the utility of changes in biomarkers over time for HF risk. We discuss studies of biomarkers to guide management and assess the efficacy of prevention strategies and multi-biomarker and multimodality approaches to improve risk prediction.
Collapse
Affiliation(s)
| | - Vijay Nambi
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX 77030, USA
| | - Biykem Bozkurt
- Department of Medicine, Cardiology Section, Winters Center for Heart Failure Research, Cardiovascular Research Institute, Baylor College of Medicine, DeBakey Veterans Affairs Medical Center, Houston, TX 77030, USA;
| |
Collapse
|
33
|
Newman JD, Anthopolos R, Ruggles KV, Cornwell M, Reynolds HR, Bangalore S, Mavromatis K, Held C, Wallentin L, Kullo IJ, McManus B, Newby LKK, Rosenberg Y, Hochman JS, Maron DJ, Berger JS. Biomarkers and cardiovascular events in patients with stable coronary disease in the ISCHEMIA Trials. Am Heart J 2023; 266:61-73. [PMID: 37604357 PMCID: PMC10843480 DOI: 10.1016/j.ahj.2023.08.007] [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: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/23/2023]
Abstract
IMPORTANCE Biomarkers may improve prediction of cardiovascular events for patients with stable coronary artery disease (CAD), but their importance in addition to clinical tests of inducible ischemia and CAD severity is unknown. OBJECTIVES To evaluate the prognostic value of multiple biomarkers in stable outpatients with obstructive CAD and moderate or severe inducible ischemia. DESIGN AND SETTING The ISCHEMIA and ISCHEMIA CKD trials randomized 5,956 participants with CAD to invasive or conservative management from July 2012 to January 2018; 1,064 participated in the biorepository. MAIN OUTCOME MEASURES Primary outcome was cardiovascular death, myocardial infarction (MI), or hospitalization for unstable angina, heart failure, or resuscitated cardiac arrest. Secondary outcome was cardiovascular death or MI. Improvements in prediction were assessed by cause-specific hazard ratios (HR) and area under the receiver operating characteristics curve (AUC) for an interquartile increase in each biomarker, controlling for other biomarkers, in a base clinical model of risk factors, left ventricular ejection fraction (LVEF) and ischemia severity. Secondary analyses were performed among patients in whom core-lab confirmed severity of CAD was ascertained by computed cardiac tomographic angiography (CCTA). EXPOSURES Baseline levels of interleukin-6 (IL-6), high sensitivity troponin T (hsTnT), growth differentiation factor 15 (GDF-15), N-terminal pro-B-type natriuretic peptide (NT-proBNP), lipoprotein a (Lp[a]), high sensitivity C-reactive protein (hsCRP), Cystatin C, soluble CD 40 ligand (sCD40L), myeloperoxidase (MPO), and matrix metalloproteinase 3 (MMP3). RESULTS Among 757 biorepository participants, median (IQR) follow-up was 3 (2-5) years, age was 67 (61-72) years, and 144 (19%) were female; 508 had severity of CAD by CCTA available. In an adjusted multimarker model with hsTnT, GDF-15, NT-proBNP and sCD40L, the adjusted HR for the primary outcome per interquartile increase in each biomarker was 1.58 (95% CI 1.22, 2.205), 1.60 (95% CI 1.16, 2.20), 1.61 (95% 1.22, 2.14), and 1.46 (95% 1.12, 1.90), respectively. The adjusted multimarker model also improved prediction compared with the clinical model, increasing the AUC from 0.710 to 0.792 (P < .01) and 0.714 to 0.783 (P < .01) for the primary and secondary outcomes, respectively. Similar findings were observed after adjusting for core-lab confirmed atherosclerosis severity. CONCLUSIONS AND RELEVANCE Among ISCHEMIA biorepository participants, biomarkers of myocyte injury/distension, inflammation, and platelet activity improved cardiovascular event prediction in addition to risk factors, LVEF, and assessments of ischemia and atherosclerosis severity. These biomarkers may improve risk stratification for patients with stable CAD.
Collapse
Affiliation(s)
- Jonathan D Newman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY.
| | - Rebecca Anthopolos
- Division of Biostatistics, Department of Population Health, NYU Langone Health, New York, NY
| | - Kelly V Ruggles
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | | | | | - Sripal Bangalore
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Kreton Mavromatis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Iftikar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Bruce McManus
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - L Kristin K Newby
- Division of Cardiology, Department of Medicine, Duke Clinical Research Institute, Durham, NC
| | - Yves Rosenberg
- Division of Cardiovascular Sciences, National Health Lung and Blood Institute, National Institute of Health, Bethesda, MD
| | - Judith S Hochman
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - David J Maron
- Department of Medicine, Stanford University, Stanford, CA
| | - Jeffrey S Berger
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
34
|
You J, Guo Y, Zhang Y, Kang JJ, Wang LB, Feng JF, Cheng W, Yu JT. Plasma proteomic profiles predict individual future health risk. Nat Commun 2023; 14:7817. [PMID: 38016990 PMCID: PMC10684756 DOI: 10.1038/s41467-023-43575-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Developing a single-domain assay to identify individuals at high risk of future events is a priority for multi-disease and mortality prevention. By training a neural network, we developed a disease/mortality-specific proteomic risk score (ProRS) based on 1461 Olink plasma proteins measured in 52,006 UK Biobank participants. This integrative score markedly stratified the risk for 45 common conditions, including infectious, hematological, endocrine, psychiatric, neurological, sensory, circulatory, respiratory, digestive, cutaneous, musculoskeletal, and genitourinary diseases, cancers, and mortality. The discriminations witnessed high accuracies achieved by ProRS for 10 endpoints (e.g., cancer, dementia, and death), with C-indexes exceeding 0.80. Notably, ProRS produced much better or equivalent predictive performance than established clinical indicators for almost all endpoints. Incorporating clinical predictors with ProRS enhanced predictive power for most endpoints, but this combination only exhibited limited improvement when compared to ProRS alone. Some proteins, e.g., GDF15, exhibited important discriminative values for various diseases. We also showed that the good discriminative performance observed could be largely translated into practical clinical utility. Taken together, proteomic profiles may serve as a replacement for complex laboratory tests or clinical measures to refine the comprehensive risk assessments of multiple diseases and mortalities simultaneously. Our models were internally validated in the UK Biobank; thus, further independent external validations are necessary to confirm our findings before application in clinical settings.
Collapse
Affiliation(s)
- Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lin-Bo Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Zhangjiang Fudan International Innovation Center, Shanghai, China.
- School of Data Science, Fudan University, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Zhou Y, Wang X, Yuan H, Wu L, Zhang B, Chen X, Zhang Y. Impact of recombinant human brain natriuretic peptide on emergency dialysis and prognosis in end-stage renal disease patients with type 4 cardiorenal syndrome. Sci Rep 2023; 13:20752. [PMID: 38007545 PMCID: PMC10676370 DOI: 10.1038/s41598-023-48125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023] Open
Abstract
Recombinant human brain natriuretic peptide (rhBNP) effects on type 4 cardiorenal syndrome (CRS) and adverse events such as heart failure rehospitalization and all-cause mortality have not been assessed in large-scale research. This study evaluated the impact of rhBNP on emergency dialysis and prognosis in end-stage renal disease (ESRD) patients with type 4 CRS, and the risk factors of emergency dialysis. This retrospective cohort study included patients with type 4 CRS and ESRD admitted for decompensated heart failure between January 2016 and December 2021. Patients were divided into the rhBNP and non-rhBNP cohorts, according to whether they were prescribed rhBNP. The primary outcomes were emergency dialysis at first admission and cardiovascular events within a month after discharge. A total of 77 patients were included in the rhBNP cohort (49 males and 28 females, median age 67) and 79 in the non-rhBNP cohort (47 males and 32 females, median age 68). After adjusting for age, residual renal function, and primary diseases, Cox regression analysis showed that rhBNP was associated with emergency dialysis (HR = 0.633, 95% CI 0.420-0.953) and cardiovascular events (HR = 0.410, 95% CI 0.159-0.958). In addition, multivariate logistic regression analysis showed that estimated glomerular filtration rate (eGFR) (OR = 0.782, 95% CI 0.667-0.917, P = 0.002) and procalcitonin (PCT) levels (OR = 1.788, 95% CI 1.193-2.680, P = 0.005) at the first visit were independent risk factors for emergency dialysis while using rhBNP was a protective factor for emergency dialysis (OR = 0.195, 95% CI 0.084-0.451, P < 0.001). This study suggests that RhBNP can improve cardiac function and reduce the occurrence of emergency dialysis and cardiovascular events in ESRD patients with type 4 CRS.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaojian Wang
- Department of Nephrology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - Hongbo Yuan
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Linke Wu
- Department of Respiratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Bin Zhang
- Department of Cardiology, Nanjing Yuhua Hospital, Nanjing, 210039, China
| | - Xiaoxia Chen
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yafeng Zhang
- Department of Public Health, Affiliated Hospital of Jiangsu University, Zhenjiang, 212003, China.
| |
Collapse
|
36
|
Riccardi M, Myhre PL, Zelniker TA, Metra M, Januzzi JL, Inciardi RM. Soluble ST2 in Heart Failure: A Clinical Role beyond B-Type Natriuretic Peptide. J Cardiovasc Dev Dis 2023; 10:468. [PMID: 37998526 PMCID: PMC10672197 DOI: 10.3390/jcdd10110468] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
Soluble (s)ST2 has been proposed as a useful biomarker for heart failure (HF) patient management. Myocardial damage or mechanical stress stimulate sST2 release. ST2 competes with a membrane bound receptor (ST2 ligand, or ST2L) for interleukin-33 (IL-33) binding, inhibiting the effects induced by the ST2L/IL-33 interaction so that excessive sST2 may contribute to myocardial fibrosis and ventricular remodeling. Compared to natriuretic peptides (NPs), sST2 concentration is not substantially affected by age, sex, body mass index, kidney function, atrial fibrillation, anemia, or HF etiology, and has low intra-individual variation. Its prognostic role as an independent marker is well reported in the literature. However, there is a gap on its use in combination with NPs, currently the only biomarkers recommended by European and American guidelines for HF management. Reflecting the activation of two distinct biological systems, a benefit from the use of sST2 and NP in combination is advocated. The aim of this review is to report the current scientific knowledge on sST2 in the acute and chronic HF settings with a particular attention to its additive role to natriuretic peptides (NPs).
Collapse
Affiliation(s)
- Mauro Riccardi
- Institute of Cardiology, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, 25121 Brescia, Italy; (M.R.); (M.M.)
| | - Peder L. Myhre
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, 1478 Nordbyhagen, Norway;
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, University of Oslo, 0313 Oslo, Norway
| | - Thomas A. Zelniker
- Department of Internal Medicine II, Division of Cardiology, Center of Cardiovascular Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, 25121 Brescia, Italy; (M.R.); (M.M.)
| | - James L. Januzzi
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, and Baim Institute for Clinical Research, Boston, MA 02215, USA;
| | - Riccardo M. Inciardi
- Institute of Cardiology, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, 25121 Brescia, Italy; (M.R.); (M.M.)
| |
Collapse
|
37
|
Salvioli S, Basile MS, Bencivenga L, Carrino S, Conte M, Damanti S, De Lorenzo R, Fiorenzato E, Gialluisi A, Ingannato A, Antonini A, Baldini N, Capri M, Cenci S, Iacoviello L, Nacmias B, Olivieri F, Rengo G, Querini PR, Lattanzio F. Biomarkers of aging in frailty and age-associated disorders: State of the art and future perspective. Ageing Res Rev 2023; 91:102044. [PMID: 37647997 DOI: 10.1016/j.arr.2023.102044] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
According to the Geroscience concept that organismal aging and age-associated diseases share the same basic molecular mechanisms, the identification of biomarkers of age that can efficiently classify people as biologically older (or younger) than their chronological (i.e. calendar) age is becoming of paramount importance. These people will be in fact at higher (or lower) risk for many different age-associated diseases, including cardiovascular diseases, neurodegeneration, cancer, etc. In turn, patients suffering from these diseases are biologically older than healthy age-matched individuals. Many biomarkers that correlate with age have been described so far. The aim of the present review is to discuss the usefulness of some of these biomarkers (especially soluble, circulating ones) in order to identify frail patients, possibly before the appearance of clinical symptoms, as well as patients at risk for age-associated diseases. An overview of selected biomarkers will be discussed in this regard, in particular we will focus on biomarkers related to metabolic stress response, inflammation, and cell death (in particular in neurodegeneration), all phenomena connected to inflammaging (chronic, low-grade, age-associated inflammation). In the second part of the review, next-generation markers such as extracellular vesicles and their cargos, epigenetic markers and gut microbiota composition, will be discussed. Since recent progresses in omics techniques have allowed an exponential increase in the production of laboratory data also in the field of biomarkers of age, making it difficult to extract biological meaning from the huge mass of available data, Artificial Intelligence (AI) approaches will be discussed as an increasingly important strategy for extracting knowledge from raw data and providing practitioners with actionable information to treat patients.
Collapse
Affiliation(s)
- Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Sara Carrino
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Eleonora Fiorenzato
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), Department of Neurosciences, University of Padova, Padova, Italy
| | - Nicola Baldini
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, Telese Terme, Italy
| | | | | |
Collapse
|
38
|
Ma H, Zhou J, Zhang M, Shen C, Jiang Z, Zhang T, Gao F. The Diagnostic Accuracy of N-Terminal Pro-B-Type Natriuretic Peptide and Soluble ST2 for Heart Failure in Chronic Kidney Disease Patients: A Comparative Analysis. Med Sci Monit 2023; 29:e940641. [PMID: 37667469 PMCID: PMC10492504 DOI: 10.12659/msm.940641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/13/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND N-terminal proatrial natriuretic peptide (NT-proBNP) levels are often markedly elevated in patients with chronic kidney disease (CKD). Identifying novel biomarkers is an important step toward effective diagnosis. Interleukin-1 receptor-like 1 (IL1RL1) protein and human/Soluble suppression of tumorigenesis-2 (sST2) are promising biomarkers for heart failure (HF). This study aimed to assess the trend of NT-proBNP and sST2 in chronic kidney disease and their diagnostic value for HF. MATERIAL AND METHODS This study was carried out on 420 patients who were divided into a no heart failure group (N=182) and a heart failure group (N=238). Spearman correlation analysis was used to test the association of sST2 and NT-proBNP with renal function. The diagnostic value of each biomarker was assessed using receiver operating characteristic (ROC) curves according to 3 different forms: Total group (n=420), non-CKD group (n=217), and CKD group (n=203). RESULTS A striking correlation between eGFR and NT-proBNP (r=-0.525; P<0.001) seemed to be far stronger than that with sST2 (r=-0.147; P<0.05). The optimum cutoff points for sST2 and NT-proBNP to detect HF were 28.960 ng/mL and 1280 pg/mL, respectively, in total, 28.71 ng/mL and 481 pg/mL, respectively, in non-CKD patients, and 30.55 ng/mL and 3314 pg/mL, respectively, in CKD patients. The combined model of sST2 and NT-proBNP was superior to the model of sST2 or NT-proBNP alone, and the difference was statistically significant (P<0.05). CONCLUSIONS The diagnostic value of sST2 is less affected by decreased renal function. sST2 combined with NT-proBNP may improve the diagnostic accuracy of HF.
Collapse
Affiliation(s)
- Hongzhen Ma
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jun Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Meng Zhang
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Chun Shen
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Zhifan Jiang
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Tao Zhang
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Fei Gao
- Department of Geriatric Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| |
Collapse
|
39
|
Nopp S, Moik F, Kraler S, Englisch C, Preusser M, von Eckardstein A, Pabinger I, Lüscher TF, Ay C. Growth differentiation factor-15 and prediction of cancer-associated thrombosis and mortality: a prospective cohort study. J Thromb Haemost 2023; 21:2461-2472. [PMID: 37192696 DOI: 10.1016/j.jtha.2023.04.043] [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: 03/31/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Patients with cancer are at increased risk of venous thromboembolism (VTE) and arterial thromboembolic/thrombotic events (ATEs). Growth differentiation factor-15 (GDF-15) improves cardiovascular risk assessment, but its predictive utility in patients with cancer remains undefined. OBJECTIVES To investigate the association of GDF-15 with the risks of VTE, ATE, and mortality in patients with cancer and its predictive utility alongside established models. METHODS The Vienna Cancer and Thrombosis Study (CATS)-a prospective, observational cohort study of patients with newly diagnosed or recurrent cancer-which was followed for 2 years, served as the study framework. Serum GDF-15 levels at study inclusion were measured, and any association with VTE, ATE, and death was determined using competing risk (VTE/ATE) or Cox regression (death) modeling. The added value of GDF-15 to established VTE risk prediction models was assessed using the Khorana and Vienna CATScore. RESULTS Among 1531 included patients with cancer (median age, 62 years; 53% men), median GDF-15 levels were 1004 ng/L (IQR, 654-1750). Increasing levels of GDF-15 were associated with the increased risks of VTE, ATE, and all-cause death ([subdistribution] hazard ratio per doubling, 1.16 [95% CI, 1.03-1.32], 1.30 [95% CI, 1.11-1.53], and 1.57 [95% CI, 1.46-1.69], respectively). After adjustment for clinically relevant covariates, the association only prevailed for all-cause death (hazard ratio, 1.21; 95% CI, 1.10-1.33) and GDF-15 did not improve the performance of the Khorana or Vienna CATScore. CONCLUSION GDF-15 is strongly associated with survival in patients with cancer, independent of the established risk factors. While an association with ATE and VTE was identified in univariable analysis, GDF-15 was not independently associated with these outcomes and failed to improve established VTE prediction models.
Collapse
Affiliation(s)
- Stephan Nopp
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Florian Moik
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria; Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria. http://www.twitter.com/FlorianMoik
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland. http://www.twitter.com/KralerSimon
| | - Cornelia Englisch
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Ingrid Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland; Royal Brompton and Harefield Hospitals and Imperial College, London, UK; School of Cardiovascular Medicine and Sciences, King's College London, London, UK. http://www.twitter.com/TomLuscher
| | - Cihan Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
40
|
Farag NM, Mousa M, Elsayed E, Ismeil A. GDF-15 and hepcidin as a therapeutic target for anemia in chronic kidney disease. Ital J Pediatr 2023; 49:106. [PMID: 37649102 PMCID: PMC10469522 DOI: 10.1186/s13052-023-01505-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Anaemia is a common presenting feature among patients with chronic kidney disease (CKD) and associated with poor clinical outcomes. We evaluated the diagnostic validity of growth differentiation factor-15 (GDF-15) and hepcidin as it is not clear if they are useful as a biomarkers of anaemia among non-dialysis CKD egyptian patients. METHOD An analytical cross-sectional study was conducted among non-dialysis CKD patients (n = 60) and apparently healthy controls (n = 28) at Minia University maternity & children Hospital. Serum levels of GDF-15 and hepcidin were determined. Predictive logistic regression models were built and post estimation receiver operator characteristics were determined to evaluate diagnostic validity of hepcidin and GDF-15 for iron deficiency anaemia. RESULTS Hepcidin and GDF-15 are significantly higher in cases than control p value (0.047 < 0.0001) respectively. The predictive value of diagnosing anaemia among CKD patients using hepcidin and GDF-15 was 72.0%, 70.0%. There was a weak negative correlation between hepcidin levels and glomerular filtration rate GFR (r = -.175, p = 0.105) in CKD patients, and significant correlation between serum GDF-15 and haemoglobin (r = -0.897, p < 0.0001), ferritin (r = 0.489, P < 0.000), Iron (r = -0.314, P = 0.002), CRP (r = 0.409, P < 0.0001). CONCLUSION Hepcidin and GDF-15 is a potential biomarker for predicting anaemia connected with inflammation among CKD Egyptian patients.
Collapse
Affiliation(s)
| | | | - Eman Elsayed
- Clinical Pathology, Minia University, Minia, Egypt
| | | |
Collapse
|
41
|
Januzzi JL, Mohebi R, Liu Y, Sattar N, Heerspink HJL, Tefera E, Vaduganathan M, Butler J, Yavin Y, Li J, Pollock CA, Perkovic V, Neal B, Hansen MK. Cardiorenal Biomarkers, Canagliflozin, and Outcomes in Diabetic Kidney Disease: The CREDENCE Trial. Circulation 2023; 148:651-660. [PMID: 37603600 DOI: 10.1161/circulationaha.123.065251] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND People with type 2 diabetes and albuminuria are at an elevated risk for cardiac and renal events. The optimal biomarkers to aid disease prediction and to understand the benefits of sodium-glucose cotransporter-2 inhibition remain unclear. METHODS Among 2627 study participants in the CREDENCE trial (Canagliflozin and Renal Events in Diabetes With Established Nephropathy Clinical Evaluation), concentrations of NT-proBNP (N-terminal pro-B-type natriuretic peptide), high-sensitivity cardiac troponin T, growth differentiation factor-15, and IGFBP7 (insulin-like growth factor binding protein 7) were measured. The effect of canagliflozin on biomarker concentrations was evaluated. The prognostic potential of each biomarker on the primary outcome (a composite of end-stage kidney disease [dialysis, transplantation, or a sustained estimated glomerular filtration rate of <15 mL·min-1·1.73 m-2], doubling of the serum creatinine level, or renal death or cardiovascular death) was assessed. RESULTS The median (quartiles 1 and 3) concentration of each biomarker was generally elevated: NT-proBNP, 180 ng/L (82, 442 ng/L); high-sensitivity cardiac troponin T, 19 ng/L (12, 29 ng/L); growth differentiation factor-15, 2595 ng/L (1852, 3775 ng/L); and IGFBP7, 121.8 ng/mL (105.4, 141.5 ng/mL). At 1 year, the biomarkers all rose by 6% to 29% in the placebo arm but only by 3% to 10% in the canagliflozin arm (all P<0.01 in multivariable linear mixed-effect models). Baseline concentrations of each biomarker were strongly predictive of cardiac and renal outcomes. When the biomarkers were analyzed together in a multimarker panel, individuals with high risk scores (hazard ratio [HR], 4.01 [95% CI, 2.52-6.35]) and moderate risk scores (HR, 2.39 [95% CI, 1.48-3.87]) showed a higher risk for the primary outcome compared with those with low risk scores. By 1 year, a 50% increase in NT-proBNP (HR, 1.11 [95% CI, 1.08-1.15]), high-sensitivity cardiac troponin T (HR, 1.86 [95% CI, 1.64-2.10]), growth differentiation factor-15 (HR, 1.45 [95% CI, 1.24-1.70]), and IGFBP7 (HR, 3.76 [95% CI, 2.54-5.56]) was associated with risk of the primary outcome. CONCLUSIONS Multiple cardiorenal stress biomarkers are strongly prognostic in people with type 2 diabetes and albuminuria. Canagliflozin modestly reduced the longitudinal trajectory of rise in each biomarker. Change in the biomarker level in addition to the baseline level augments the primary outcome prediction. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT02065791.
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (J.L.J., R.M., Y.L.)
- Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, MA (J.L.J.)
| | - Reza Mohebi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (J.L.J., R.M., Y.L.)
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (J.L.J., R.M., Y.L.)
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, UK (N.S.)
| | - Hiddo J L Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, the Netherlands (H.J.L.H.)
| | - Eshetu Tefera
- Janssen Research & Development, LLC, Spring House, PA (E.T., Y.Y., M.K.H.)
| | - Muthiah Vaduganathan
- Cardiology Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.V.)
| | - Javed Butler
- University of Mississippi Medical Center, Jackson (J.B.)
- Baylor Scott & White Institute, Dallas, TX (J.B.)
| | - Yshai Yavin
- Janssen Research & Development, LLC, Spring House, PA (E.T., Y.Y., M.K.H.)
| | - Jingwei Li
- Department of Cardiology, People's Liberation Army General Hospital, Beijing, China (J.L.)
- Department of Cardiology, Xinqiao Hospital, Army Military Medical University, Chongqing, China (J.L.)
- The George Institute for Global Health (J.L., V.P., B.N.), UNSW Sydney, Australia
| | - Carol A Pollock
- Kolling Institute, Royal North Shore Hospital University of Sydney, NSW, Australia (C.A.P.)
| | - Vlado Perkovic
- The George Institute for Global Health (J.L., V.P., B.N.), UNSW Sydney, Australia
- Faculty of Medicine (V.P.), UNSW Sydney, Australia
- The Royal North Shore Hospital, Sydney, Australia (V.P.)
| | - Bruce Neal
- The George Institute for Global Health (J.L., V.P., B.N.), UNSW Sydney, Australia
- Charles Perkins Centre, University of Sydney, NSW, Australia (B.N.)
- Imperial College London, UK (B.N.)
| | - Michael K Hansen
- Janssen Research & Development, LLC, Spring House, PA (E.T., Y.Y., M.K.H.)
| |
Collapse
|
42
|
Ullah A, Sajid S, Qureshi M, Kamran M, Anwaar MA, Naseem MA, Zaman MU, Mahmood F, Rehman A, Shehryar A, Nadeem MA. Novel Biomarkers and the Multiple-Marker Approach in Early Detection, Prognosis, and Risk Stratification of Cardiac Diseases: A Narrative Review. Cureus 2023; 15:e42081. [PMID: 37602073 PMCID: PMC10434821 DOI: 10.7759/cureus.42081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Cardiac diseases are a primary cause of mortality worldwide, underscoring the importance of early identification and risk stratification to enhance patient outcomes. Biomarkers have become important tools for the risk assessment of cardiovascular disease and monitoring disease progression. This narrative review focuses on the multiple-marker approach, which involves simultaneously evaluating several biomarkers for the early detection and risk stratification of heart diseases. The review covers the clinical applications of novel biomarkers, such as high-sensitivity troponin, galectin-3, source of tumorigenicity 2, B-type natriuretic peptide and N-terminal pro-B-type natriuretic peptide, growth differentiation factor 15, myeloperoxidase, fatty acid-binding protein, C-reactive protein, lipoprotein-associated phospholipase A2, microRNAs, circulating endothelial cells, and ischemia-modified albumin. These biomarkers have demonstrated potential in identifying people who are at high risk for developing heart disease and in providing prognostic data. Given the complexity of cardiac illnesses, the multiple-marker approach to risk assessment is extremely beneficial. Implementing the multiple-marker strategy can improve risk stratification, diagnostic accuracy, and patient care in heart disease patients.
Collapse
Affiliation(s)
| | - Samar Sajid
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Maria Qureshi
- Family Medicine, Ayub Medical College, Abbottabad, PAK
| | | | - Mohammad Ahsan Anwaar
- Internal Medicine, CMH Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | | | | | - Fizza Mahmood
- Cardiology/Cardiac Surgery, Shifa International Hospital Islamabad, Islamabad, PAK
| | | | | | - Muhammad A Nadeem
- Medicine and Surgery, Shifa International Hospital Islamabad, Islamabad, PAK
| |
Collapse
|
43
|
Conte M, De Feo MS, Frantellizzi V, Di Rocco A, Farcomeni A, De Cristofaro F, Maria R, Pisani AR, Rubini G, De Vincentis G. Sex differences in 123I-mIBG scintigraphy imaging techniques in patients with heart failure. Expert Rev Med Devices 2023; 20:769-778. [PMID: 37466442 DOI: 10.1080/17434440.2023.2239139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/07/2023] [Accepted: 07/18/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND 123I-mIBG-scintigraphy could be a useful stratifying tool for patients with heart failure (HF). The purpose of this retrospective study is to evaluate whether there are differences between men and women with HF in terms of the prediction of cardiac arrhythmic events (AE). RESEARCH AND METHODS A total of 306 patients, before implantable-cardioverter-defibrillator (ICD) implantation, were evaluated. They underwent 123I-mIBG-scintigraphy and an evaluation of the results was performed after 85 months of follow-up. Early and late planar and SPECT cardiac images were acquired. Heart-to-mediastinum ratio (HM) for planar images and the sum of the segmental scores (SS) for SPECT were calculated. RESULTS In the general population, age, early SS (ESS), late SS (LSS), and ejection fraction (EF) were statistically significant for the prediction of AE at Cox regression, while early and late HM (eHM,lHM) were not significative for the prediction of AE. Population was divided into females and males and univariate analysis was conducted separately for the two cohorts: no significant variables for prediction of AE were found in females. For males, ESS, LSS, EF, and late HM were statistically significant predictors of AE. The overall survival was similar in males and females, but the risk of AE is lower in males than in females. CONCLUSIONS 123I-mIBG represents a more effective tool for the prediction of AE in male patients than in women.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| | - Arianna Di Rocco
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| | - Alessio Farcomeni
- Department of Economics & Finance, University of Rome "Tor Vergata", Rome, Italy
| | - Flaminia De Cristofaro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| | - Ricci Maria
- Nuclear Medicine Unit, Cardarelli Hospital, Campobasso, Italy
| | | | - Giuseppe Rubini
- Nuclear Medicine Department, University of Bari "Aldo Moro", Bari, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza, "Sapienza" University of Rome, Rome Italy
| |
Collapse
|
44
|
Petersen TB, de Bakker M, Asselbergs FW, Harakalova M, Akkerhuis KM, Brugts JJ, van Ramshorst J, Lumbers RT, Ostroff RM, Katsikis PD, van der Spek PJ, Umans VA, Boersma E, Rizopoulos D, Kardys I. HFrEF subphenotypes based on 4210 repeatedly measured circulating proteins are driven by different biological mechanisms. EBioMedicine 2023; 93:104655. [PMID: 37327673 DOI: 10.1016/j.ebiom.2023.104655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND HFrEF is a heterogenous condition with high mortality. We used serial assessments of 4210 circulating proteins to identify distinct novel protein-based HFrEF subphenotypes and to investigate underlying dynamic biological mechanisms. Herewith we aimed to gain pathophysiological insights and fuel opportunities for personalised treatment. METHODS In 382 patients, we performed trimonthly blood sampling during a median follow-up of 2.1 [IQR:1.1-2.6] years. We selected all baseline samples and two samples closest to the primary endpoint (PEP; composite of cardiovascular mortality, HF hospitalization, LVAD implantation, and heart transplantation) or censoring, and applied an aptamer-based multiplex proteomic approach. Using unsupervised machine learning methods, we derived clusters from 4210 repeatedly measured proteomic biomarkers. Sets of proteins that drove cluster allocation were analysed via an enrichment analysis. Differences in clinical characteristics and PEP occurrence were evaluated. FINDINGS We identified four subphenotypes with different protein profiles, prognosis and clinical characteristics, including age (median [IQR] for subphenotypes 1-4, respectively:70 [64, 76], 68 [60, 79], 57 [47, 65], 59 [56, 66]years), EF (30 [26, 36], 26 [20, 38], 26 [22, 32], 33 [28, 37]%), and chronic renal failure (45%, 65%, 36%, 37%). Subphenotype allocation was driven by subsets of proteins associated with various biological functions, such as oxidative stress, inflammation and extracellular matrix organisation. Clinical characteristics of the subphenotypes were aligned with these associations. Subphenotypes 2 and 3 had the worst prognosis compared to subphenotype 1 (adjHR (95%CI):3.43 (1.76-6.69), and 2.88 (1.37-6.03), respectively). INTERPRETATION Four circulating-protein based subphenotypes are present in HFrEF, which are driven by varying combinations of protein subsets, and have different clinical characteristics and prognosis. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01851538https://clinicaltrials.gov/ct2/show/NCT01851538. FUNDING EU/EFPIA IMI2JU BigData@Heart grant n°116074, Jaap Schouten Foundation and Noordwest Academie.
Collapse
Affiliation(s)
- Teun B Petersen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands; Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Marie de Bakker
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Health Data Research UK and Institute of Health Informatics, University College London, Gower St, London, United Kingdom
| | - Magdalena Harakalova
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, University of Utrecht, Heidelberglaan 100, Utrecht, the Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Jasper J Brugts
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Wilhelminalaan 12, Alkmaar, the Netherlands
| | - R Thomas Lumbers
- British Heart Foundation Research Accelerator, University College London, Gower St, London, UK; Institute of Health Informatics, University College London, Gower St, London, UK; Health Data Research UK London, University College London, Gower St, London, UK
| | | | - Peter D Katsikis
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Peter J van der Spek
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Victor A Umans
- Department of Cardiology, Northwest Clinics, Wilhelminalaan 12, Alkmaar, the Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, Rotterdam, the Netherlands.
| |
Collapse
|
45
|
Dolivo DM, Reed CR, Gargiulo KA, Rodrigues AE, Galiano RD, Mustoe TA, Hong SJ. Anti-fibrotic effects of statin drugs: a review of evidence and mechanisms. Biochem Pharmacol 2023:115644. [PMID: 37321414 DOI: 10.1016/j.bcp.2023.115644] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
Fibrosis is a pathological repair process common among organs, that responds to damage by replacement of tissue with non-functional connective tissue. Despite the widespread prevalence of tissue fibrosis, manifesting in numerous disease states across myriad organs, therapeutic modalities to prevent or alleviate fibrosis are severely lacking in quantity and efficacy. Alongside development of new drugs, repurposing of existing drugs may be a complementary strategy to elect anti-fibrotic compounds for pharmacologic treatment of tissue fibrosis. Drug repurposing can provide key advantages to de novo drug discovery, harnessing the benefits of previously elucidated mechanisms of action and already existing pharmacokinetic profiles. One class of drugs a wealth of clinical data and extensively studied safety profiles is the statins, a class of antilipidemic drugs widely prescribed for hypercholesterolemia. In addition to these widely utilized lipid-lowering effects, increasing data from cellular, pre-clinical mammalian, and clinical human studies have also demonstrated that statins are able to alleviate tissue fibrosis originating from a variety of pathological insults via lesser-studied, pleiotropic effects of these drugs. Here we review literature demonstrating evidence for direct effects of statins antagonistic to fibrosis, as well as much of the available mechanistic data underlying these effects. A more complete understanding of the anti-fibrotic effects of statins may enable a clearer picture of their anti-fibrotic potential for various clinical indications. Additionally, more lucid comprehension of the mechanisms by which statins exert anti-fibrotic effects may aid in development of novel therapeutic agents that target similar pathways but with greater specificity or efficacy.
Collapse
Affiliation(s)
- David M Dolivo
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States.
| | - Charlotte R Reed
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Kristine A Gargiulo
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Adrian E Rodrigues
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Robert D Galiano
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Thomas A Mustoe
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Seok Jong Hong
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States.
| |
Collapse
|
46
|
Abstract
Cardiovascular disease (CVD) is prevalent in patients with chronic kidney disease (CKD) and it is responsible for approximately half of all CKD-related deaths. CVDs are the primary cause of death in hemodialysis patients due to major adverse cardiovascular events. Therefore, better approaches for differentiating chronic hemodialysis patients at higher cardiovascular risk will help physicians improve clinical outcomes. Hence, there is an urgent need to discover feasible and reliable cardiac biomarkers to improve diagnostic accuracy, reflect myocardial injury, and identify high-risk patients. Numerous biomarkers that have significant prognostic value with respect to adverse CVD outcomes in the setting of mild to severe CKD have been identified. Therefore, a better understanding of the positive clinical impact of cardiac biomarkers on CVD patient outcomes is an important step toward prevention and improving treatment in the future. In this review, we address the relationship between cardiovascular biomarkers and CKD treatment strategies to elucidate the underlying importance of these biomarkers to patient outcomes.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung, Taiwan
| | - Chih-Chieh Chen
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Tze-Kiong Er
- Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
47
|
Girerd N, Levy D, Duarte K, Ferreira JP, Ballantyne C, Collier T, Pizard A, Björkman J, Butler J, Clark A, Cleland JG, Delles C, Diez J, González A, Hazebroek M, Ho J, Huby AC, Hwang SJ, Latini R, Mariottoni B, Mebazaa A, Pellicori P, Sattar N, Sever P, Staessen JA, Verdonschot J, Heymans S, Rossignol P, Zannad F. Protein Biomarkers of New-Onset Heart Failure: Insights From the Heart Omics and Ageing Cohort, the Atherosclerosis Risk in Communities Study, and the Framingham Heart Study. Circ Heart Fail 2023; 16:e009694. [PMID: 37192292 DOI: 10.1161/circheartfailure.122.009694] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 03/03/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND We sought to identify protein biomarkers of new-onset heart failure (HF) in 3 independent cohorts (HOMAGE cohort [Heart Omics and Ageing], ARIC study [Atherosclerosis Risk in Communities], and FHS [Framingham Heart Study]) and assess if and to what extent they improve HF risk prediction compared to clinical risk factors alone. METHODS A nested case-control design was used with cases (incident HF) and controls (without HF) matched on age and sex within each cohort. Plasma concentrations of 276 proteins were measured at baseline in ARIC (250 cases/250 controls), FHS (191/191), and HOMAGE cohort (562/871). RESULTS In single protein analysis, after adjusting for matching variables and clinical risk factors (and correcting for multiple testing), 62 proteins were associated with incident HF in ARIC, 16 in FHS, and 116 in HOMAGE cohort. Proteins associated with incident HF in all cohorts were BNP (brain natriuretic peptide), NT-proBNP (N-terminal pro-B-type natriuretic peptide), eukaryotic translation initiation factor 4E-BP1 (4E-binding protein 1), hepatocyte growth factor (HGF), Gal-9 (galectin-9), TGF-alpha (transforming growth factor alpha), THBS2 (thrombospondin-2), and U-PAR (urokinase plasminogen activator surface receptor). The increment in C-index for incident HF based on a multiprotein biomarker approach, in addition to clinical risk factors and NT-proBNP, was 11.1% (7.5%-14.7%) in ARIC, 5.9% (2.6%-9.2%) in FHS, and 7.5% (5.4%-9.5%) in HOMAGE cohort, all P<0.001), each of which was a larger increase than that for NT-proBNP on top of clinical risk factors. Complex network analysis revealed a number of overrepresented pathways related to inflammation (eg, tumor necrosis factor and interleukin) and remodeling (eg, extracellular matrix and apoptosis). CONCLUSIONS A multiprotein biomarker approach improves prediction of incident HF when added to natriuretic peptides and clinical risk factors.
Collapse
Affiliation(s)
- Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| | - Daniel Levy
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (D.L., J.H., S.-J.H)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (D.L., J.H., S.-J.H.)
| | - Kevin Duarte
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| | - Joao Pedro Ferreira
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| | | | - Timothy Collier
- London School of Hygiene and Tropical Medicine, United Kingdom (T.C.)
| | - Anne Pizard
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
- Inserm 1024, Institut de Biologie de l'École Normale Supérieure (IBENS), PSL University of Paris, France (A.P.)
| | | | - Javed Butler
- Department of Medicine, University of Mississippi School of Medicine, Jackson (J.B.)
| | - Andrew Clark
- Hull York Medical School, Castle Hill Hospital, Cottingham, United Kingdom (A.C.)
| | - John G Cleland
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health and Wellbeing, University of Glasgow, United Kingdom (J.G.C., P.P.)
- National Heart and Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, United Kingdom (J.G.C.)
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (C.D.C.)
| | - Javier Diez
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain (J.D.C., A.G.C.)
- CIBERCV, Carlos III Institute of Health, Madrid, Spain (J.D.C., A.G.C.)
| | - Arantxa González
- CIBERCV, Carlos III Institute of Health, Madrid, Spain (J.D.C., A.G.C.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain (J.D.C., A.G.C.)
- Departments of Nephrology, and Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain (A.G.C.)
| | - Mark Hazebroek
- Department of Cardiology, Maastricht University Medical Centre, Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, the Netherlands (M.H.C.)
| | - Jennifer Ho
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (D.L., J.H., S.-J.H)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (D.L., J.H., S.-J.H.)
| | - Anne-Cécile Huby
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA (D.L., J.H., S.-J.H)
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD (D.L., J.H., S.-J.H.)
| | - Roberto Latini
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy (R.L.)
| | | | - Alexandre Mebazaa
- UMRS 942; University Paris Diderot; APHP, University Hospitals Saint Louis Lariboisière, France (A.M.)
| | - Pierpaolo Pellicori
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health and Wellbeing, University of Glasgow, United Kingdom (J.G.C., P.P.)
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (N.S.)
| | - Peter Sever
- International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London, United Kingdom (P.S.)
| | - Jan A Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium (J.A.S.)
| | - Job Verdonschot
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium (J.A.S.)
| | - Stephane Heymans
- Department of Cardiovascular Research, University of Leuven, UZ Herestraat, Belgium (S.H.)
- Netherlands Heart Institute (ICIN), Utrecht, the Netherlands (S.H.)
- Department of Clinical Genetics, Maastricht University Medical Center, the Netherlands (S.H.)
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques- Plurithématique 14-33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.G., K.D., J.P.F., A.P., A.-C.H., P.R., F.Z.)
| |
Collapse
|
48
|
Ruban SM, Ramadass K, Singh G, Talapaneni SN, Kamalakar G, Gadipelly CR, Mannepalli LK, Sugi Y, Vinu A. Organocatalysis with carbon nitrides. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2188879. [PMID: 37007670 PMCID: PMC10054243 DOI: 10.1080/14686996.2023.2188879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Carbon nitrides, a distinguished class of metal-free catalytic materials, have presented a good potential for chemical transformations and are expected to become prominent materials for organocatalysis. This is largely possible due to their low cost, exceptional thermal and chemical stability, non-toxicity, ease of functionalization, porosity development, etc. Especially, the carbon nitrides with increased porosity and nitrogen contents are more versatile than their bulk counterparts for catalysis. These N-rich carbon nitrides are discussed in the earlier parts of the review. Later, the review highlights the role of such carbon nitride materials for the various organic catalytic reactions including Knoevenagel condensation, oxidation, hydrogenation, esterification, transesterification, cycloaddition, and hydrolysis. The recently emerging concepts in carbon nitride-based organocatalysis have been given special attention. In each of the sections, the structure-property relationship of the materials was discussed and related to their catalysis action. Relevant comparisons with other catalytic materials are also discussed to realize their real potential value. The perspective, challenges, and future directions are also discussed. The overall objective of this review is to provide up-to-date information on new developments in carbon nitride-based organic catalysis reactions that could see them rising as prominent catalytic materials in the future.
Collapse
Affiliation(s)
- Sujanya Maria Ruban
- Global Innovative Centre for Advanced Nanomaterials (GICAN), College of Engineering, Science and Environment (CESE), School of Engineering, The University of Newcastle, Callaghan, Australia
| | - Kavitha Ramadass
- Global Innovative Centre for Advanced Nanomaterials (GICAN), College of Engineering, Science and Environment (CESE), School of Engineering, The University of Newcastle, Callaghan, Australia
| | - Gurwinder Singh
- Global Innovative Centre for Advanced Nanomaterials (GICAN), College of Engineering, Science and Environment (CESE), School of Engineering, The University of Newcastle, Callaghan, Australia
| | | | - Gunda Kamalakar
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | | | | | - Yoshihiro Sugi
- Global Innovative Centre for Advanced Nanomaterials (GICAN), College of Engineering, Science and Environment (CESE), School of Engineering, The University of Newcastle, Callaghan, Australia
- Faculty of Engineering, Gifu University, Gifu, Japan
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials (GICAN), College of Engineering, Science and Environment (CESE), School of Engineering, The University of Newcastle, Callaghan, Australia
| |
Collapse
|
49
|
Bakhshi H, Michelhaugh SA, Bruce SA, Seliger SL, Qian X, Ambale Venkatesh B, Varadarajan V, Bagchi P, Lima JAC, deFilippi C. Association between proteomic biomarkers and myocardial fibrosis measured by MRI: the multi-ethnic study of atherosclerosis. EBioMedicine 2023; 90:104490. [PMID: 36857966 PMCID: PMC10006438 DOI: 10.1016/j.ebiom.2023.104490] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Cardiac magnetic resonance imaging (CMR) determines the extent of interstitial fibrosis, measured by increased extracellular volume (ECV), and replacement fibrosis with late gadolinium myocardial enhancement (LGE). Despite advances in detection, the pathophysiology of subclinical myocardial fibrosis is incompletely understood. Targeted proteomic discovery technologies enable quantification of low abundance circulating proteins to elucidate cardiac fibrosis mechanisms. METHODS Using a cross-sectional design, we selected 92 LGE+ cases and 92 LGE- demographically matched controls from the Multi-Ethnic Study of Atherosclerosis. Similarly, we selected 156 cases from the highest ECV quartile and matched with 156 cases from the lowest quartile. The plasma serum proteome was analyzed using proximity extension assays to determine differential regulation of 92 proteins previously implicated with cardiovascular disease. Results were analyzed using volcano plots of statistical significance vs. magnitude of change and Bayesian additive regression tree (BART) models to determine importance. FINDINGS After adjusting for false discovery, higher ECV was significantly associated with 17 proteins. Using BART, Plasminogen activator inhibitor 1, Insulin-like growth factor-binding protein 1, and N-terminal pro-B-type natriuretic peptide were associated with higher ECV after accounting for other proteins and traditional cardiovascular risk factors. In contrast, no circulating proteins were associated with replacement fibrosis. INTERPRETATIONS Our results suggest unique circulating proteomic signatures associated with interstitial fibrosis emphasizing its systemic influences. With future validation, protein panels may identify patients who may develop interstitial fibrosis with progression to heart failure. FUNDING This research was supported by contracts and grants from NHLBI, NCATS and the Inova Heart and Vascular Institute.
Collapse
Affiliation(s)
- Hooman Bakhshi
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Scott A Bruce
- Department of Statistics, Texas A&M University, College Station, TX, USA
| | - Stephen L Seliger
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Geriatric Research and Education Clinical Center, VA Maryland Healthcare System, Baltimore, MD, USA
| | - Xiaoxiao Qian
- Inova Heart and Vascular Institute, Falls Church, VA, USA
| | | | | | - Pramita Bagchi
- Department of Statistics, George Mason University, Fairfax, VA, USA
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
50
|
deFilippi CR, Tran H, Gattani R, Daniels LB, Shah P, Ilkhanoff L, Christenson R, Lima JA, Seliger S. Association of cardiac troponin T and growth differentiation factor 15 with replacement and interstitial cardiac fibrosis in community dwelling adults: The multi-ethnic study of atherosclerosis. Front Cardiovasc Med 2023; 10:1104715. [PMID: 36844723 PMCID: PMC9949377 DOI: 10.3389/fcvm.2023.1104715] [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: 12/07/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Background Subclinical abnormalities in myocardial structure (stage B heart failure) may be identified by cardiac and non-organ specific biomarkers. The associations of high-sensitivity cardiac troponin T (hs-cTnT) and growth differentiation factor-15 (GDF-15) with cardiac magnetic resonance imaging (CMR) interstitial fibrosis (extracellular volume [ECV]) is unknown and for GDF-15 the association with replacement (late gadolinium enhancement [LGE]) is also unknown. GDF-15 is a systemic biomarker also released by myocytes associated with fibrosis and inflammation. We sought to define the associations of hs-cTnT and GDF-15 with these CMR fibrosis measures in the MESA cohort. Methods We measured hs-cTnT and GDF-15 in MESA participants free of cardiovascular disease at exam 5. CMR measurements were complete in 1737 for LGE and 1258 for ECV assessment. We estimated the association of each biomarker with LGE and increased ECV (4th quartile) using logistic regression, adjusted for demographics and risk factors. Results Mean age of the participants was 68 ± 9 years. Unadjusted, both biomarkers were associated with LGE, but after adjustment only hs-cTnT concentrations remained significant (4th vs. 1st quartile OR] 7.5, 95% CI: 2.1, 26.6). For interstitial fibrosis both biomarkers were associated with 4th quartile ECV, but the association was attenuated compared to replacement fibrosis. After adjustment, only hs-cTnT concentrations remained significant (1st to 4th quartile OR 1.7, 95%CI: 1.1, 2.8). Conclusion Our findings identify that both interstitial and replacement fibrosis are associated with myocyte cell death/injury, but GDF-15 a non-organ specific biomarker prognostic for incident cardiovascular disease is not associated with preclinical evidence of cardiac fibrosis.
Collapse
Affiliation(s)
- Christopher R. deFilippi
- Inova Heart and Vascular Institute, Falls Church, VA, United States,*Correspondence: Christopher R. deFilippi,
| | - Henry Tran
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | - Raghav Gattani
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | - Lori B. Daniels
- Division of Cardiology, University of California and San Diego Medical Center, San Diego, CA, United States
| | - Palak Shah
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | | | - Robert Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joao A. Lima
- The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Stephen Seliger
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|