1
|
Lin M, Guo J, Gu Z, Tang W, Tao H, You S, Jia D, Sun Y, Jia P. Machine learning and multi-omics integration: advancing cardiovascular translational research and clinical practice. J Transl Med 2025; 23:388. [PMID: 40176068 DOI: 10.1186/s12967-025-06425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The global burden of cardiovascular diseases continues to rise, making their prevention, diagnosis and treatment increasingly critical. With advancements and breakthroughs in omics technologies such as high-throughput sequencing, multi-omics approaches can offer a closer reflection of the complex physiological and pathological changes in the body from a molecular perspective, providing new microscopic insights into cardiovascular diseases research. However, due to the vast volume and complexity of data, accurately describing, utilising, and translating these biomedical data demands substantial effort. Researchers and clinicians are actively developing artificial intelligence (AI) methods for data-driven knowledge discovery and causal inference using various omics data. These AI approaches, integrated with multi-omics research, have shown promising outcomes in cardiovascular studies. In this review, we outline the methods for integrating machine learning, one of the most successful applications of AI, with omics data and summarise representative AI models developed that leverage various omics data to facilitate the exploration of cardiovascular diseases from underlying mechanisms to clinical practice. Particular emphasis is placed on the effectiveness of using AI to extract potential molecular information to address current knowledge gaps. We discuss the challenges and opportunities of integrating omics with AI into routine diagnostic and therapeutic practices and anticipate the future development of novel AI models for wider application in the field of cardiovascular diseases.
Collapse
Affiliation(s)
- Mingzhi Lin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhilin Gu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Wenyi Tang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hongqian Tao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Shilong You
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
2
|
Xie R, Vlaski T, Sha S, Brenner H, Schöttker B. Sex-specific proteomic signatures improve cardiovascular risk prediction for the general population without cardiovascular disease or diabetes. J Adv Res 2025:S2090-1232(25)00194-8. [PMID: 40154735 DOI: 10.1016/j.jare.2025.03.034] [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: 01/11/2025] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
INTRODUCTION Accurate prediction of 10-year major adverse cardiovascular events (MACE) is critical for effective disease prevention and management. Although the SCORE2 model introduced sex-specific algorithms, opportunities remain to further refine prediction. OBJECTIVES To evaluate whether adding sex-specific proteomic profiles to the SCORE2 model enhances 10-year MACE risk prediction in the large UK Biobank (UKB) cohort. METHODS Data from 47,382 UKB participants, aged 40 to 69 years without prior cardiovascular disease or diabetes, were utilized. Proteomic profiling of plasma samples was conducted using the Olink Explore 3072 platform, measuring 2,923 unique proteins, of which 2,085 could be used. Sex-specific Least Absolute Shrinkage and Selection Operator (LASSO) regression was used for biomarker selection. Model performance was assessed by changes in Harrell's C-index (a measure of discrimination), net reclassification index (NRI), and integrated discrimination index (IDI). RESULTS During 10-year follow-up, 2,163 participants experienced MACE. Overall, 18 proteins were selected by LASSO regression, with 5 of them identified in both sexes, 7 only in males, and 6 only in females. Incorporating these proteins significantly improved the C-index of the SCORE2 model from 0.713 to 0.778 (P < 0.001) in the total population. The improvement was greater in males (C-index increase from 0.684 to 0.771; Δ = +0.087) than in females (from 0.720 to 0.769; Δ = +0.049). The WAP four-disulfide core domain protein (WFDC2) and the growth/differentiation factor 15 (GDF15) were the proteins contributing the strongest C-index increase in both sexes, even more than the N-terminal prohormone of brain natriuretic peptide (NTproBNP). CONCLUSION The derived sex-specific 10-year MACE risk prediction models, combining 12 protein concentrations among men and 11 protein concentrations among women with the SCORE2 model, significantly improved the discriminative abilities of the SCORE2 model. This study shows the potential of sex-specific proteomic profiles for enhanced cardiovascular risk stratification and personalized prevention strategies.
Collapse
Affiliation(s)
- Ruijie Xie
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69115 Heidelberg, Germany
| | - Tomislav Vlaski
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69115 Heidelberg, Germany
| | - Sha Sha
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| |
Collapse
|
3
|
Makram OM, Nain P, Vasbinder A, Weintraub NL, Guha A. Cardiovascular Risk Assessment and Prevention in Cardio-Oncology: Beyond Traditional Risk Factors. Cardiol Clin 2025; 43:1-11. [PMID: 39551552 DOI: 10.1016/j.ccl.2024.08.003] [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] [Indexed: 11/19/2024]
Abstract
This review goes beyond traditional approaches in cardio-oncology, highlighting often-neglected factors impacting patient care. Social determinants, environment, health care access, and gut microbiome significantly influence patient outcomes. Powerful tools like multi-omics and wearable technologies offer deeper insights into real-world experiences. The future lies in integrating these advancements with established practices to achieve precision cardio-oncology care. By crafting tailored therapies and continuously updating comprehensive management plans based on real-time data, we can unlock the full potential of personalized care for all patients.
Collapse
Affiliation(s)
- Omar M Makram
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; Department of Medicine, Cardio-Oncology Program, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Priyanshu Nain
- Department of Medicine, Cardio-Oncology Program, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA; Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Alexi Vasbinder
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, WA, USA
| | - Neal L Weintraub
- Department of Medicine, Cardio-Oncology Program, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA; Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Avirup Guha
- Department of Medicine, Cardio-Oncology Program, Cardiology Division, Medical College of Georgia at Augusta University, Augusta, GA, USA; Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
4
|
Bankier S, Talukdar H, Khan M, Mocci G, Sukhavasi K, Hao K, Ma A, Ruusalepp A, Schadt EE, Kovacic JC, Michoel T, Björkegren JL. Plasma proteins are integral to gene-regulatory networks acting within and across blood cells, the arterial wall and major metabolic organs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.22.25320723. [PMID: 39973987 PMCID: PMC11839005 DOI: 10.1101/2025.01.22.25320723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The plasma proteome is the future for diagnostic markers for common diseases, like cardiometabolic disorders (CMDs) and coronary artery disease (CAD). The reliability of these markers requires identifying their source-organ(s). We profiled 974 plasma proteins in 532 CAD-patients of the STARNET study with arterial wall, major metabolic organ, and blood transcriptomic data. 144 plasma cis-pQTLs colocalized with tissue eQTLs including eight CMD/CAD GWAS genes. 262 plasma proteins correlated with their corresponding tissue "seed" genes whereof 101 in the liver. 851 plasma proteins were strongly associated with the activity of gene-regulatory networks (GRNs), particularly cross-tissue GRNs. The Adipose-Liver Plasma LeptIN-regulating GRN78 with the top key driver UCHL1 in fat stood out. Beyond genetics, explaining up to 20% of plasma protein variation, and a limited number of mostly hepatic seed genes, plasma proteins are integral to GRNs acting within and across blood cells, the arterial wall and major metabolic organs.
Collapse
Affiliation(s)
- Sean Bankier
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Husain Talukdar
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Mariyam Khan
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Giuseppe Mocci
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Ke Hao
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Angela Ma
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Arno Ruusalepp
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
- Clinical Gene Networks AB, Stockholm, Sweden
| | - Eric E Schadt
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York 10029, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
- St. Vincent's Clinical School, University of NSW, Sydney, Australia
| | - Tom Michoel
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Johan Lm Björkegren
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Clinical Gene Networks AB, Stockholm, Sweden
| |
Collapse
|
5
|
Kjaergaard AD, Vaag A, Jensen VH, Olsen MH, Højlund K, Vestergaard P, Hansen T, Thomsen RW, Jessen N. YKL-40, cardiovascular events, and mortality in individuals recently diagnosed with type 2 diabetes: A Danish cohort study. Diabetes Res Clin Pract 2025; 219:111970. [PMID: 39719182 DOI: 10.1016/j.diabres.2024.111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
AIMS We investigated the association of the inflammatory biomarker YKL-40 with cardiovascular events (CVEs) and mortality in individuals with type 2 diabetes. METHODS We followed 11,346 individuals recently diagnosed with type 2 diabetes for up to 14 years. Baseline YKL-40 levels (measured in 9,010 individuals) were grouped into percentiles (0-33 %, 34-66 %, 67-90 %, and 91-100 %) and analyzed continuously (per 1 SD log increment), with comparisons to CRP (measured in 9,644 individuals). Cox regression assessed associations with atrial fibrillation (AF), ischemic stroke (IS), venous thromboembolism (VTE), myocardial infarction (MI), heart failure (HF), peripheral artery disease (PAD), and all-cause, cardiovascular, and cancer mortality. RESULTS Adjusted HRs (95% CIs) for the highest (91-100%) versus the lowest (0-33%) YKL-40 percentile category were 1.31 (1.04-1.66) for AF, 1.43 (0.98-2.07) for IS, 1.07 (0.65-1.76) VTE, 0.88 (0.52-1.48) for MI, 1.66 (1.19-2.31) for HF, 1.66 (1.12-2.48) for PAD, and 2.18 (1.85-2.56) for all-cause, 1.64 (1.07-2.50) for cardiovascular, and 2.73 (2.05-3.63) for cancer mortality. Each 1 SD log increase in YKL-40 and CRP levels similarly increased CVE risks, with CRP being superior for MI and cardiovascular mortality. CONCLUSIONS YKL-40 is a prognostic biomarker for most CVEs, and even more so for all-cause mortality, primarily driven by cancer-related causes.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
| | - Verena H Jensen
- Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
| | - Michael H Olsen
- Department of Internal Medicine and Steno Diabetes Center Zealand, Holbæk Hospital, Holbæk, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Peter Vestergaard
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Reimar W Thomsen
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
6
|
Xu H, Wen Y, Zheng H, Jiang D, Chen W. Allergic disease and keratoconus: A two-sample univariable and multivariable Mendelian randomization study. World Allergy Organ J 2024; 17:100993. [PMID: 39650195 PMCID: PMC11621933 DOI: 10.1016/j.waojou.2024.100993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 12/11/2024] Open
Abstract
Background There is accumulating evidence that allergy is a risk factor for keratoconus. Nonetheless the association between allergic disease and keratoconus remains controversial. We performed a two-sample Mendelian randomization (MR) study to determine the putative causal association of 4 allergic diseases (allergic conjunctivitis, allergic asthma, allergic rhinitis and atopic dermatitis) with keratoconus. Methods Summary statistics were obtained from genome-wide association studies (GWAS) of allergic conjunctivitis (AC) (20,958 cases and 356,319 controls), allergic asthma (AA) (9631 cases and 210,122 controls), allergic rhinitis (AR) (11,009 cases and 359,149 controls), atopic dermatitis (AD) (13,473 cases and 336,589 controls), keratoconus (KC) (2116 cases and 24,626 controls) and 91 circulating inflammatory cytokines (n = 14,824). Two-sample univariable and multivariable MR analyses were performed. A two-step MR was then applied to determine whether systemic inflammatory cytokines mediated the effect of allergic disease on keratoconus. Results The causal odds ratio (OR) estimate of genetically determined KC was 1.66 (95% CI: 1.32-2.08; P < 0.001) for AC, 1.29 (95% CI: 1.10-1.51, P = 0.0014) for AA, 1.39 (95% CI: 1.15-1.68; P < 0.001) for AR and 1.30 (95% CI: 1.17-1.45, P < 0.001) for AD. Multivariable MR indicated a suggestive association between AC and KC after conditioning on other allergic diseases (OR 1.61; 95% CI: 1.10-2.34; P adjusted = 0.054). Two-step MR revealed that the effect was not mediated by systemic inflammatory cytokines. Conclusions Our findings provide evidence of a potential causal relationship between AC and KC. The effect of AC on KC may be mediated via other systemic inflammatory cytokines not included in the present study, or by alternative mechanisms. These findings may offer insight for prevention and intervention strategies to lower the risk of KC in patients with AC.
Collapse
Affiliation(s)
- Hanlu Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yajing Wen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Huikang Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Dan Jiang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
7
|
Li M, Li M, Wang Z, Zhang Y. The Combined Effect of the Systemic Immune-Inflammation Index and Aortic Valve Calcification on Major Adverse Cardiovascular Events in Patients with Coronary Heart Disease. J Inflamm Res 2024; 17:8375-8384. [PMID: 39529998 PMCID: PMC11552382 DOI: 10.2147/jir.s493735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background The combined effect of systemic immune-inflammation index (SII) and aortic valve calcification (AVC) on the risk of major adverse cardiovascular events (MACE) in patients with coronary heart disease (CHD) remains unclear. This study aimed to investigate their combined association with MACE in CHD. Methods This retrospective cohort study included 846 CHD patients. SII was calculated as platelet count × neutrophil count / lymphocyte count, and AVC status was determined by echocardiography. Patients were divided into four groups based on median SII and AVC presence: Low SII + AVC (-), High SII + AVC (-), Low SII + AVC (+), and High SII + AVC (+). Cox regression, subgroup and sensitivity analyses assessed the association between SII + AVC and MACE. Results Multivariate Cox regression revealed that, compared to the Low SII + AVC (-), MACE risk increased 6.542-fold in the High SII + AVC (+) group and 1.605-fold in the High SII + AVC (-) group (P < 0.05). Subgroup analysis indicated that, compared to the Low SII + AVC (-), MACE risk was significantly elevated in the High SII + AVC (-) group for patients over 60, both genders, with hypertension, hyperlipidemia, or without diabetes (P < 0.05). In the Low SII + AVC (+) group, MACE risk was elevated only in males (P < 0.05). The High SII + AVC (+) group had increased MACE risk in all subgroups except those with diabetes (P < 0.05). After excluding patients with estimated glomerular filtration rate < 60 mL/min/1.73m², the high SII + AVC (+) group remained significantly associated with increased MACE risk (P = 0.001), as did the High SII + AVC (-) group (P = 0.031). Conclusion The combination of SII and AVC is significantly associated with MACE risk in patients with CHD.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Cardiology, Chest Hospital of Zhengzhou University, Zhengzhou, 450008, People’s Republic of China
- Department of Cardiology, Henan Province Chest Hospital, Zhengzhou, 450008, People’s Republic of China
| | - Mengchun Li
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People’s Republic of China
| | - Yongbo Zhang
- Department of Cardiology, Chest Hospital of Zhengzhou University, Zhengzhou, 450008, People’s Republic of China
- Department of Cardiology, Henan Province Chest Hospital, Zhengzhou, 450008, People’s Republic of China
| |
Collapse
|
8
|
Stewart RAH, Robledo KP, Tonkin AM, Keech A, Kritharides L, Marschner I, Janus E, Thompson PL, Watts GF, Zeller T, White HD, Simes J. Plasma Protein Biomarkers and Long-Term Cardiovascular Mortality Risk in Patients With Chronic Coronary Heart Disease. J Am Heart Assoc 2024; 13:e034367. [PMID: 39450716 PMCID: PMC11935700 DOI: 10.1161/jaha.123.034367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Protein biomarkers that reflect different pathophysiological pathways have been associated with the risk of adverse cardiovascular events. However, it is uncertain whether these associations are sustained with increasing years after the biomarkers are measured. METHODS AND RESULTS In this cohort study, 7745 patients with coronary heart disease who participated in the LIPID (Long-Term Intervention With Pravastatin in Ischemic Disease) trial, BNP (B-type natriuretic peptide), troponin I, cystatin-C, C-reactive protein, d-dimer and midregional proadrenomedullin were measured at baseline and after 1 year. Discrimination of plasma biomarker concentrations for cardiovascular death were evaluated in landmark analyses from 1 year for the next 5 years of the randomized trial, and for 10 additional years after trial completion. All 6 biomarkers were associated with risk of cardiovascular death (n=1903) both during and after the clinical trial (each P<0.001). C-statistics for BNP were 0.706 and 0.704; cystatin-C, 0.686 and 0.693; troponin I, 0.686 and 0.689; C-reactive protein, 0.655 and 0.684; d-dimer, 0.670 and 0.679, and midregional adrenomedullin, 0.686 and 0.688, respectively. In multivariable models, adding all 6 biomarkers to models with clinical risk factors increased the C-statistic for cardiovascular death from 0.709 to 0.775 during the clinical trial, and from 0.713 to 0.751 during 10-year follow-up after the randomized trial (P<0.001 for both). CONCLUSIONS In patients with chronic coronary heart disease, biomarkers that reflect different pathophysiological pathways are associated with the risk of cardiovascular death for at least the next 15 years.
Collapse
Affiliation(s)
- Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora—Health New ZealandAucklandNew Zealand
| | - Kristy P. Robledo
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Andrew M. Tonkin
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVICAustralia
| | - Anthony Keech
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Leonard Kritharides
- Department of Cardiology, Concord HospitalThe University of SydneyConcordNSWAustralia
- ANZAC Medical Research Institute, Faculty of Medicine, University of SydneyConcordNSWAustralia
| | - Ian Marschner
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| | - Edward Janus
- Western Health Chronic Disease Alliance and Department of Medicine, Western Health—Melbourne Medical SchoolUniversity of MelbourneParkvilleVic3010Australia
| | - Peter L. Thompson
- School of Population and Global HealthThe University of Western AustraliaPerthWAAustralia
| | - Gerald F. Watts
- Medical SchoolThe University of Western AustraliaPerthWAAustralia
| | - Tanja Zeller
- University Heart Centre HamburgHamburgGermany
- Department of General and Interventional Cardiology, German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Campus ResearchHamburgGermany
| | - Harvey D. White
- Green Lane Cardiovascular Service, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora—Health New ZealandAucklandNew Zealand
| | - John Simes
- Faculty Medicine and Health, NHMRC Clinical Trials CentreUniversity of Syndey and The Royal Prince Alfred HospitalCamperdownNSWAustralia
| |
Collapse
|
9
|
Royer P, Björnson E, Adiels M, Josefson R, Hagberg E, Gummesson A, Bergström G. Large-scale plasma proteomics in the UK Biobank modestly improves prediction of major cardiovascular events in a population without previous cardiovascular disease. Eur J Prev Cardiol 2024; 31:1681-1689. [PMID: 38546334 DOI: 10.1093/eurjpc/zwae124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/24/2024] [Indexed: 10/11/2024]
Abstract
AIMS Improved identification of individuals at high risk of developing cardiovascular disease would enable targeted interventions and potentially lead to reductions in mortality and morbidity. Our aim was to determine whether use of large-scale proteomics improves prediction of cardiovascular events beyond traditional risk factors (TRFs). METHODS AND RESULTS Using proximity extension assays, 2919 plasma proteins were measured in 38 380 participants of the UK Biobank. Both data- and literature-based feature selection and trained models using extreme gradient boosting machine learning were used to predict risk of major cardiovascular events (MACEs: fatal and non-fatal myocardial infarction, stroke, and coronary artery revascularization) during a 10-year follow-up. Area under the curve (AUC) and net reclassification index (NRI) were used to evaluate the additive value of selected protein panels to MACE prediction by Systematic COronary Risk Evaluation 2 (SCORE2) or the 10 TRFs used in SCORE2. SCORE2 and SCORE2 refitted to UK Biobank data predicted MACE with AUCs of 0.740 and 0.749, respectively. Data-driven selection identified 114 proteins of greatest relevance for prediction. Prediction of MACE was not improved by using these proteins alone (AUC of 0.758) but was significantly improved by combining these proteins with SCORE2 or the 10 TRFs (AUC = 0.771, P < 001, NRI = 0.140, and AUC = 0.767, P = 0.03, NRI 0.053, respectively). Literature-based protein selection (113 proteins from five previous studies) also improved risk prediction beyond TRFs while a random selection of 114 proteins did not. CONCLUSION Large-scale plasma proteomics with data-driven and literature-based protein selection modestly improves prediction of future MACE beyond TRFs.
Collapse
Affiliation(s)
- Patrick Royer
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Critical Care, University Hospital of Martinique, Fort-de-France, Martinique, French West Indies, France
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
| | - Martin Adiels
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rebecca Josefson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
| | - Eva Hagberg
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, Institute of Medicine, Gothenburg University, PO Box 100,405 30 Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
10
|
Liu S, Qian F, Lu Q, Deng Y, Qu W, Lin X, Li R, Li R, Guo T, Pan A, Liu G. Association of life's essential 8 with risk of recurrent cardiovascular events among patients with coronary heart disease. Int J Cardiol 2024; 412:132318. [PMID: 38971538 DOI: 10.1016/j.ijcard.2024.132318] [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: 03/19/2024] [Revised: 06/17/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
AIMS To examine the association of Life's Essential 8 (LE8) with the risk of recurrent cardiovascular events among patients with CHD. METHODS This prospective cohort study included 11,997 patients with CHD from the UK Biobank. The LE8 score was generated using five lifestyle factors (diet, body mass index, physical activity, smoking, and sleep) and three biological factors (blood lipids, blood glucose, and blood pressure). LE8 score ranged from 0 to 100 and was categorized into quartiles. Cox proportional hazards regression models were applied to estimate the hazard ratio (HR) and 95% CI (confidence interval). RESULTS During a median follow up of 12.5 years, we documented 3366 recurrent cardiovascular events, 1068 myocardial infarction, 1829 heart failure events, 703 strokes, and 934 cardiovascular deaths. The multivariable-adjusted HR (95% CI) for the highest versus the lowest quartile of LE8 score was 0.57 (0.50, 0.65) for recurrent cardiovascular events, 0.66 (0.52, 0.83) for myocardial infarction, 0.54 (0.45, 0.67) for heart failure, 0.50 (0.36, 0.68) for stroke, and 0.46 (0.37, 0.56) for cardiovascular death. Furthermore, the population attributable fraction of the lowest to the highest quartile of LE8 score were ranged from 16.2% to 32.5% for the various cardiovascular outcomes. In addition, biomarkers including renal function and inflammation collectively explained 47.6%-87.7% of the associations between the lifestyle factors and recurrent cardiovascular events. CONCLUSIONS Better cardiovascular health as measured by LE8 was associated with significantly lower risk of recurrent cardiovascular events among patients with CHD. Clinicians should prioritize educating patients with CHD on the importance of optimal cardiovascular health for secondary prevention. In addition, our findings indicated significant mediation effect of biomarkers involving of glycemic control, renal function, liver function, lipid profile, and systemic inflammation on the associations between overall lifestyle factors and recurrent cardiovascular events.
Collapse
Affiliation(s)
- Sen Liu
- Nanchang Center for Disease Control and Prevention, Nanchang, China; Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Frank Qian
- Section of Cardiovascular Medicine, Boston Medical Center, and Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Qi Lu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yulei Deng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wensheng Qu
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruyi Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Lab of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Qu Z, Lu Y, Ran Y, Xu D, Guo Z, Cheng M. Chitinase‑3 like‑protein‑1: A potential predictor of cardiovascular disease (Review). Mol Med Rep 2024; 30:176. [PMID: 39129301 PMCID: PMC11332322 DOI: 10.3892/mmr.2024.13300] [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: 05/27/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024] Open
Abstract
Chitinase‑3 like‑protein‑1 (CHI3L1), a glycoprotein belonging to the glycoside hydrolase family 18, binds to chitin; however, this protein lacks chitinase activity. Although CHI3L1 is not an enzyme capable of degrading chitin, it plays significant roles in abnormal glucose and lipid metabolism, indicating its involvement in metabolic disorders. In addition, CHI3L1 is considered a key player in inflammatory diseases, with clinical data suggesting its potential as a predictor of cardiovascular disease. CHI3L1 regulates the inflammatory response of various cell types, including macrophages, vascular smooth muscle cells and fibroblasts. In addition, CHI3L1 participates in vascular remodeling and fibrosis, contributing to the pathogenesis of cardiovascular disease. At present, research is focused on elucidating the role of CHI3L1 in cardiovascular disease. The present systematic review was conducted to comprehensively evaluate the effects of CHI3L1 on cardiovascular cells, and determine the potential implications in the occurrence and progression of cardiovascular disease. The present study may further the understanding of the involvement of CHI3L1 in cardiovascular pathology, demonstrating its potential as a therapeutic target or biomarker in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Zhuojian Qu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Yirui Lu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Yutong Ran
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Donghua Xu
- Central Laboratory of The First Affiliated Hospital, Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Zhiliang Guo
- Department of Spine Surgery, The 80th Group Army Hospital of Chinese PLA, Weifang, Shandong 261021, P.R. China
| | - Min Cheng
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
12
|
Luo H, Petrera A, Hauck SM, Rathmann W, Herder C, Gieger C, Hoyer A, Peters A, Thorand B. Association of plasma proteomics with mortality in individuals with and without type 2 diabetes: Results from two population-based KORA cohort studies. BMC Med 2024; 22:420. [PMID: 39334377 PMCID: PMC11438072 DOI: 10.1186/s12916-024-03636-0] [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: 04/03/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Protein biomarkers may contribute to the identification of vulnerable subgroups for premature mortality. This study aimed to investigate the association of plasma proteins with all-cause and cause-specific mortality among individuals with and without baseline type 2 diabetes (T2D) and evaluate their impact on the prediction of all-cause mortality in two prospective Cooperative Health Research in the Region of Augsburg (KORA) studies. METHODS The discovery cohort comprised 1545 participants (median follow-up 15.6 years; 244 with T2D: 116 total, 62 cardiovascular, 31 cancer-related and 23 other-cause deaths; 1301 without T2D: 321 total, 114 cardiovascular, 120 cancer-related and 87 other-cause deaths). The validation cohort comprised 1031 participants (median follow-up 6.9 years; 203 with T2D: 76 total, 45 cardiovascular, 19 cancer-related and 12 other-cause deaths; 828 without T2D: 169 total, 74 cardiovascular, 39 cancer-related and 56 other-cause deaths). We used Cox regression to examine associations of 233 plasma proteins with all-cause and cause-specific mortality and Lasso regression to construct prediction models for all-cause mortality stratifying by baseline T2D. C-index, category-free net reclassification index (cfNRI), and integrated discrimination improvement (IDI) were conducted to evaluate the predictive performance of built prediction models. RESULTS Thirty-five and 62 proteins, with 29 overlapping, were positively associated with all-cause mortality in the group with and without T2D, respectively. Out of these, in the group with T2D, 35, eight, and 26 were positively associated with cardiovascular, cancer-related, and other-cause mortality, while in the group without T2D, 55, 41, and 47 were positively associated with respective cause-specific outcomes in the pooled analysis of both cohorts. Regulation of insulin-like growth factor (IGF) transport and uptake by IGF-binding proteins emerged as a unique pathway enriched for all-cause and cardiovascular mortality in individuals with T2D. The combined model containing the selected proteins (five and 12 proteins, with four overlapping, in the group with and without T2D, respectively) and clinical risk factors improved the prediction of all-cause mortality by C-index, cfNRI, and IDI. CONCLUSIONS This study uncovered shared and unique mortality-related proteins in persons with and without T2D and emphasized the role of proteins in improving the prediction of mortality in different T2D subgroups.
Collapse
Affiliation(s)
- Hong Luo
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Annika Hoyer
- Biostatistics and Medical Biometry, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany.
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany.
| |
Collapse
|
13
|
Zhang X, Zhang JK, Wu X, Liu X, Liu T, Chen KY. Predictive Value of the Naples Prognostic Score for Cardiovascular Outcomes in Patients With Chronic Kidney Disease Receiving Percutaneous Coronary Intervention. Angiology 2024:33197241285970. [PMID: 39298739 DOI: 10.1177/00033197241285970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The Naples prognostic score (NPS) is a novel multidimensional inflammatory and nutritional assessment system in cancer patients. However, its significance in patients with chronic kidney disease (CKD) after percutaneous coronary intervention (PCI) remains unclear. The study has a single-center, retrospective design and included 631 patients with CKD who underwent index PCI between 2019 and 2022. All participants were divided into 2 groups according to the NPS (Low-risk group: n = 209; High-risk group: n = 422) and followed up until November 2022. The primary endpoint was Major Adverse Cardiac Events (MACE). NPS predicted MACE events better than other scores, besides, high-risk NPS with severe renal dysfunction (RD) group (MODEL 2) had superior MACE diagnostic efficiency than NPS high-risk group lonely. (NPS: AUC: 0.605, P < .001; MODEL 2: AUC: 0.624, P < .001, respectively). Kaplan-Meier survival analysis of two groups showed that high-risk group had higher incidence of MACE (P < .001). Meanwhile, high-risk group had higher MACE events [adjusted Hazard Ratio (aHR) 2.013, 95% CI 1.294, 3.132; P = .002]. NPS is an independent prognostic factor for CKD patients undergoing index PCI before operation whose predictive value for survival prognosis is better than other nutritional and inflammatory indicators. Compared with low NPS, patients with high NPS have a relatively poor prognosis.
Collapse
Affiliation(s)
- Xue Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jing-Kun Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xue Wu
- Institute for Global Health Sciences, University of California, San Francisco, CA, USA
| | - Xing Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Kang-Yin Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Lee DY. Emerging Circulating Biomarkers for Enhanced Cardiovascular Risk Prediction. J Lipid Atheroscler 2024; 13:262-279. [PMID: 39355403 PMCID: PMC11439747 DOI: 10.12997/jla.2024.13.3.262] [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: 01/04/2024] [Revised: 04/12/2024] [Accepted: 06/06/2024] [Indexed: 10/03/2024] Open
Abstract
Cardiovascular disease (CVD) continues to be the primary cause of mortality worldwide, underscoring the importance of identifying additional cardiovascular risk factors. The consensus is that lipid levels alone do not fully reflect the status of atherosclerosis, thus necessitating extensive research on cardiovascular biomarkers. This review encompasses a wide spectrum of methodologies for identifying novel risk factors or biomarkers for CVD. Inflammation, oxidative stress, plaque instability, cardiac remodeling, and fibrosis play pivotal roles in CVD pathogenesis. We introduce and discuss several promising biomarkers-namely, osteocalcin, angiogenin, lipoprotein-associated phospholipase A2, growth differentiation factor 15, galectin-3, growth stimulation expressed gene 2, and microRNAs, all of which have potential implications in the assessment and management of cardiovascular risk.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Câmara SMA, Hochberg MC, Miller R, Ryan AS, Orwig D, Gruber-Baldini AL, Guralnik J, Magder LS, Feng Z, Falvey JR, Beamer BA, Magaziner J. Sustained IL-6 and sTNF-αR1 levels after hip fracture predict 5-year mortality: A prospective cohort study from the Baltimore Hip Studies. J Am Geriatr Soc 2024; 72:2644-2655. [PMID: 38864591 PMCID: PMC11905919 DOI: 10.1111/jgs.19018] [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: 12/20/2023] [Revised: 04/05/2024] [Accepted: 05/05/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Persistent inflammation is associated with adverse health outcomes, but its impact on mortality has not been investigated previously among hip fracture patients. This article aims to investigate the influence of changes in levels of cytokines in the 2 months after a hip fracture repair on 5-year mortality. METHODS This is a prospective cohort study from the Baltimore Hip Studies (BHS) with 191 community-dwelling older men and women (≥65 years) who had recently undergone surgical repair of an acute hip fracture, with recruitment from May 2006 to June 2011. Plasma interleukin-6 (IL-6), soluble tumor necrosis factor alpha receptor1 (sTNFα-R1), and interleukin-1 receptor agonist (IL-1RA) were obtained within 22 days of admission and at 2 months. All-cause mortality over 5 years was determined. Logistic regression analysis tested the associations between the cytokines' trajectories and mortality over 5 years, adjusted for covariates (age, sex, education, body mass index, lower extremity physical activities of daily living, and Charlson comorbidity index). RESULTS High levels of IL-6 and sTNFα-R1 at baseline with small or no decline at 2 months were associated with higher odds of 5-year mortality compared with those with lower levels at baseline and greater decline at 2 months after adjustment for age, and other potential confounders (OR = 4.71, p = 0.01 for IL-6; OR = 15.03, p = 0.002 for sTNFα-R1). Similar results that failed to reach significance were found for IL-1RA (OR = 2.40, p = 0.18). Those with higher levels of cytokines at baseline with greater decline did not have significantly greater mortality than the reference group, those with lower levels at baseline and greater decline. CONCLUSION Persistent elevation of plasma IL-6 and sTNFα-R1 levels within the first 2 months after hospital admission in patients with hip fracture is associated with higher 5-year mortality. These patients may benefit from enhanced care and earlier intensive interventions to reduce the risk of death.
Collapse
Affiliation(s)
- Saionara M A Câmara
- Department of Physiotherapy, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marc C Hochberg
- Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Medical Care Clinical Center, VA Maryland Health Care System, Baltimore, Maryland, USA
| | - Ram Miller
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Alice S Ryan
- Geriatric Research Education and Clinical Center, Veterans Affairs Maryland Healthcare System, Baltimore, Maryland, USA
- Division of Gerontology, Geriatrics, and Palliative Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Denise Orwig
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ann L Gruber-Baldini
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jack Guralnik
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Laurence S Magder
- Division of Biostatistics and Bioinformatics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhaoyong Feng
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jason R Falvey
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Physical Therapy and Rehabilitation Science, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brock A Beamer
- Geriatric Research Education and Clinical Center, Veterans Affairs Maryland Healthcare System, Baltimore, Maryland, USA
- Division of Gerontology, Geriatrics, and Palliative Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jay Magaziner
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Baragetti A, Grigore L, Olmastroni E, Mattavelli E, Catapano AL. Plasma proteins associate with carotid plaques and predict incident atherosclerotic cardiovascular events. Vascul Pharmacol 2024; 156:107394. [PMID: 38866119 DOI: 10.1016/j.vph.2024.107394] [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: 02/17/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Performing non-invasive carotid imaging is challenging, owing inter-operator variability and organizational barriers, but plasma proteomics can offer an alternative. We sought plasma proteins that associate with the presence of carotid plaques, their number and predict the incidence of clinically overt atherosclerotic cardiovascular events (ASCVD) above currently recognized risk factors in "apparently healthy" subjects. METHODS We studied the plasma levels of 368 proteins in 664 subjects from the PLIC study, who underwent an ultrasound imaging screening of the carotids to check for the presence of plaques. We clustered, by artificial intelligence (A.I.), the proteins that associate with the presence, the number of plaques and that predict incident ASCVDs over 22 years (198 events were registered). FINDINGS 299/664 subjects had at least 1 carotid plaque (1+) (77 with only one plaque, 101 with 2 plaques, 121 with ≥3 plaques (3+)). The remaining 365 subjects with no plaques acted as controls. 106 proteins were associated with 1+ plaques, but 97 proteins significantly predicted 3+ plaques only (AUC = 0.683 (0.601-0.785), p < 0.001), when considered alone. A.I. underscored 87 proteins that improved the performance of the classical risk factors both in detecting 3+ plaques (AUC = 0.918 (0.887-0.943) versus risk factors alone, AUC = 0.760 (0.716-0.801), p < 0.001) and in predicting the incident ASCVD (AUC = 0.739 (0.704-0.773) vs risk factors alone AUC = 0.559 (0.521-0.598), p < 0.001). The chemotaxis/migration of leukocytes and interleukins/cytokines signaling were biological pathways mostly represented by these proteins. DISCUSSION AND CONCLUSIONS Plasma proteomics marks the number of carotid plaques and improve the prediction of incidence ASCVDs in apparently healthy subjects.
Collapse
Affiliation(s)
- Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | | | - Elena Olmastroni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Elisa Mattavelli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy; Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy; IRCCS Multimedica Hospital, Milan, Italy
| |
Collapse
|
17
|
Yuan Y, Tian W, Wei X, Zhu Y, Liu F, Zhang X. Diagnostic and prognostic value of serum Cys-C, retinol-binding protein, and ischemia-modified albumin in patients with coronary heart disease: A diagnostic accuracy study. Medicine (Baltimore) 2024; 103:e39415. [PMID: 39213212 PMCID: PMC11365651 DOI: 10.1097/md.0000000000039415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
The use of 3 biomarkers - cystatin-C (Cys-C), retinol-binding protein (RBP), and ischemia-modified albumin (IMA) - for the clinical classification and outcome of coronary heart disease (CHD) has not been adequately evaluated. We explored the serum levels of these 3 markers and evaluated their diagnostic and prognostic values in patients with CHD. This retrospective case-control study, conducted between June 2017 and June 2018, included 201 patients with CHD hospitalized at the Henan Provincial People's Hospital and 127 healthy individuals from Henan Provincial People's Hospital as controls. Cys-C, RBP, IMA levels, and other laboratory parameters in the 2 groups were determined, and patient outcomes were analyzed. Cys-C, RBP, and IMA levels were higher in the case group than in the control group (P < .05). Logistic regression analysis confirmed that these 3 biomarkers were independent risk factors for CHD. Each indicator has clinical significance in the diagnosis and prognosis of CHD, with RBP being the most significant. The AUC value for CHD detection using a combination of the 3 indicators was 0.783, and the sensitivity and specificity values were 78% and 74.6%, respectively. Simultaneous detection of Cys-C, RBP, and IMA could be an optimal method for early diagnosis and prognosis of CHD.
Collapse
Affiliation(s)
- Youhua Yuan
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Wenqian Tian
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Xiaoxia Wei
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Ya Zhu
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Fengzhen Liu
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| | - Xiaohuan Zhang
- Department of Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, People’s Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
18
|
Sacco MA, Gualtieri S, Cordasco F, Tarallo AP, Verrina MC, Princi A, Bruni A, Garofalo E, Aquila I. The Role of Adrenomedullin as a Predictive Marker of the Risk of Death and Adverse Clinical Events: A Review of the Literature. J Clin Med 2024; 13:4847. [PMID: 39200990 PMCID: PMC11355278 DOI: 10.3390/jcm13164847] [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: 07/04/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Adrenomedullin (ADM) is a vasodilatory peptide that plays a crucial role in maintaining cardiovascular health through its various biological functions. ADM was discovered in the acidic extract of human pheochromocytoma tissue and has been recognized for its significant effects on the vascular system. The main functions of ADM include vasodilation, controlling blood pressure and maintaining vascular integrity, although its role on cardiovascular health is broader. Research has shown that elevated levels of adrenomedullin have been observed in a large number of severe diseases, with high risk of death. In this work, we examined the role of ADM as a predictive molecule of the risk of mortality and adverse clinical outcome through a narrative review of the scientific literature. The results were divided based on the pathologies and anatomical districts examined. This review demonstrates how ADM shows, in many diseases and different systems, a close correlation with the risk of mortality. These results prove the value of ADM as a prognostic marker in various clinical contexts and diseases, with utility in the stratification of the risk of clinical worsening and/or death and in the evaluation of therapeutic efficacy. The results open new perspectives with respect to the concrete possibility that ADM enters clinical practice as an effective diagnostic and prognostic marker of death as well as a molecular target for therapies aimed at patient survival.
Collapse
Affiliation(s)
- Matteo Antonio Sacco
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Saverio Gualtieri
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Fabrizio Cordasco
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Alessandro Pasquale Tarallo
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Maria Cristina Verrina
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Aurora Princi
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| | - Andrea Bruni
- Intensive Care Unit, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (A.B.); (E.G.)
| | - Eugenio Garofalo
- Intensive Care Unit, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (A.B.); (E.G.)
| | - Isabella Aquila
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, ‘Magna Graecia’ University of Catanzaro, 88100 Catanzaro, Italy; (M.A.S.); (S.G.); (F.C.); (A.P.T.); (M.C.V.); (A.P.)
| |
Collapse
|
19
|
Feng Y, He LQ. Soluble ST2: A Novel Biomarker for Diagnosis and Prognosis of Cardiovascular Disease. Curr Med Sci 2024; 44:669-679. [PMID: 39096477 DOI: 10.1007/s11596-024-2907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
The increasing incidence of cardiovascular disease (CVD) is a significant global health concern, affecting millions of individuals each year. Accurate diagnosis of acute CVD poses a formidable challenge, as misdiagnosis can significantly decrease patient survival rates. Traditional biomarkers have played a vital role in the diagnosis and prognosis of CVDs, but they can be influenced by various factors, such as age, sex, and renal function. Soluble ST2 (sST2) is a novel biomarker that is closely associated with different CVDs. Its low reference change value makes it suitable for continuous measurement, unaffected by age, kidney function, and other confounding factors, facilitating risk stratification of CVDs. Furthermore, the combination of sST2 with other biomarkers can enhance diagnostic accuracy and prognostic value. This review aims to provide a comprehensive overview of sST2, focusing on its diagnostic and prognostic value as a myocardial marker for different types of CVDs and discussing the current limitations of sST2.
Collapse
Affiliation(s)
- Yin Feng
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Qun He
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
20
|
Yu H, Wang Z, Zhu B, Jia Z, Luo J, Han X, Chen H, Shao R. A humanized Anti-YKL-40 antibody inhibits tumor development. Biochem Pharmacol 2024; 225:116335. [PMID: 38824968 DOI: 10.1016/j.bcp.2024.116335] [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: 03/12/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Drugs specifically targeting YKL-40, an over-expressed gene (CHI3L1) in various diseases remain developed. The current study is to create a humanized anti-YKL-40 neutralizing antibody and characterize its potentially therapeutic signature. We utilized in silico CDR-grafting bioinformatics to replace the complementarity determining regions (CDRs) of human IgG1 with mouse CDRs of our previously established anti-YKL-40 antibody (mAY). In fifteen candidates (VL1-3/VH1-5) of heavy and light chain variable region combination, one antibody L3H4 named Rosazumab demonstrated strong binding affinity with YKL-40 (KD = 4.645 × 10-8 M) and high homology with human IgG (80 %). In addition, we established different overlapping amino acid peptides of YKL-40 and found that Rosazumab specifically bound to residues K337, K342, and R344, the KR-rich functional domain of YKL-40. Rosazumab inhibited migration and tube formation of YKL-40-expressing tumor cells and induced tumor cell apoptosis. Mechanistically, Rosazumab induced interaction of N-cadherin with β-catenin and activation of downstream MST1/RASSF1/Histone H2B axis, leading to chromosomal DNA breakage and cell apoptosis. Treatment of xenografted tumor mice with Rosazumab twice a week for 4 weeks inhibited tumor growth and angiogenesis, but induced tumor apoptosis. Rosazumab injected in mice distributed to blood, tumor, and other multiple organs, but did not impact in function or structure of liver and kidney, indicating non-detectable toxicity in vivo. Collectively, the study is the first one to demonstrate that a humanized YKL-40 neutralizing antibody offers a valuable means to block tumor development.
Collapse
Affiliation(s)
- Haihui Yu
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziyi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bowen Zhu
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziheng Jia
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Luo
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao Han
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui Chen
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
21
|
Liu Z, Petinrin OO, Toseef M, Chen N, Wong KC. Construction of Immune Infiltration-Related LncRNA Signatures Based on Machine Learning for the Prognosis in Colon Cancer. Biochem Genet 2024; 62:1925-1952. [PMID: 37792224 DOI: 10.1007/s10528-023-10516-4] [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: 07/10/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023]
Abstract
Colon cancer is one of the malignant tumors with high morbidity, lethality, and prevalence across global human health. Molecular biomarkers play key roles in its prognosis. In particular, immune-related lncRNAs (IRL) have attracted enormous interest in diagnosis and treatment, but less is known about their potential functions. We aimed to investigate dysfunctional IRL and construct a risk model for improving the outcomes of patients. Nineteen immune cell types were collected for identifying house-keeping lncRNAs (HKLncRNA). GSE39582 and TCGA-COAD were treated as the discovery and validation datasets, respectively. Four machine learning algorithms (LASSO, Random Forest, Boruta, and Xgboost) and a Gaussian mixture model were utilized to mine the optimal combination of lncRNAs. Univariate and multivariate Cox regression was utilized to construct the risk score model. We distinguished the functional difference in an immune perspective between low- and high-risk cohorts calculated by this scoring system. Finally, we provided a nomogram. By leveraging the microarray, sequencing, and clinical data for immune cells and colon cancer patients, we identified the 221 HKLncRNAs with a low cell type-specificity index. Eighty-seven lncRNAs were up-regulated in the immune compared to cancer cells. Twelve lncRNAs were beneficial in improving performance. A risk score model with three lncRNAs (CYB561D2, LINC00638, and DANCR) was proposed with robust ROC performance on an independent dataset. According to immune-related analysis, the risk score is strongly associated with the tumor immune microenvironment. Our results emphasized IRL has the potential to be a powerful and effective therapy for enhancing the prognostic of colon cancer.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | | | - Muhammad Toseef
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Nanjun Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
Lin R, Zhu Y, Chen W, Wang Z, Wang Y, Du J. Identification of Circulating Inflammatory Proteins Associated with Calcific Aortic Valve Stenosis by Multiplex Analysis. Cardiovasc Toxicol 2024; 24:499-512. [PMID: 38589550 DOI: 10.1007/s12012-024-09854-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
Calcific aortic valve stenosis (CAVS) is characterized by increasing inflammation and progressive calcification in the aortic valve leaflets and is a major cause of death in the aging population. This study aimed to identify the inflammatory proteins involved in CAVS and provide potential therapeutic targets. We investigated the observational and causal associations of 92 inflammatory proteins, which were measured using affinity-based proteomic assays. Firstly, the case-control cohort identified differential proteins associated with the occurrence and progression of CAVS. Subsequently, we delved into exploring the causal impacts of these associated proteins through Mendelian randomization. This involved utilizing genetic instruments derived from cis-protein quantitative loci identified in genome-wide association studies, encompassing a cohort of over 400,000 individuals. Finally, we investigated the gene transcription and protein expression levels of inflammatory proteins by single-cell and immunohistochemistry analysis. Multivariate logistic regression and spearman's correlation analysis showed that five proteins showed a significant positive correlation with disease severity. Mendelian randomization showed that elevated levels of two proteins, namely, matrix metallopeptidase-1 (MMP1) and sirtuin 2 (SIRT2), were associated with an increased risk of CAVS. Immunohistochemistry and single-cell transcriptomes showed that expression levels of MMP1 and SIRT2 at the tissue and cell levels were significantly higher in calcified valves than in non-calcified control valves. These findings indicate that MMP1 and SIRT2 are causally related to CAVS and open up the possibility for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Rui Lin
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yuexin Zhu
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Weiyao Chen
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Zhiao Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yuan Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Jie Du
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
23
|
Nyárády BB, Kiss LZ, Bagyura Z, Merkely B, Dósa E, Láng O, Kőhidai L, Pállinger É. Growth and differentiation factor-15: A link between inflammaging and cardiovascular disease. Biomed Pharmacother 2024; 174:116475. [PMID: 38522236 DOI: 10.1016/j.biopha.2024.116475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
Age-related disorders are closely linked to the accumulation of senescent cells. The senescence-associated secretory phenotype (SASP) sustains and progresses chronic inflammation, which is involved in cellular and tissue dysfunction. SASP-related growth and differentiation factor-15 (GDF-15) is an immunoregulatory cytokine that is coupled to aging and thus may have a regulatory role in the development and maintenance of atherosclerosis, a major cause of cardiovascular disease (CVD). Although the effects of GDF-15 are tissue-specific and dependent on microenvironmental changes such as inflammation, available data suggest that GDF-15 has a significant role in CVD. Thus, GDF-15 is a promising biomarker and potential therapeutic target for atherosclerotic CVD.
Collapse
Affiliation(s)
- Balázs Bence Nyárády
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Loretta Zsuzsa Kiss
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Zsolt Bagyura
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Edit Dósa
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Orsolya Láng
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| |
Collapse
|
24
|
wang J, Weng H, Qian Y, Wang Y, Wang L, Wang X, Zhang P, Wang Z. The impact of serum BNP on retinal perfusion assessed by an AI-based denoising optical coherence tomography angiography in CHD patients. Heliyon 2024; 10:e29305. [PMID: 38655359 PMCID: PMC11035033 DOI: 10.1016/j.heliyon.2024.e29305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
Background To investigate the correlation between retinal vessel density (VD) parameters with serum B-type natriuretic peptide (BNP) in patients with coronary heart disease (CHD) using novel optical coherence tomography angiography (OCTA) denoising images based on artificial intelligence (AI). Methods OCTA images of the optic nerve and macular area were obtained using a Canon-HS100 OCT device in 176 patients with CHD. Baseline information and blood test results were recorded. Results Retinal VD parameters of the macular and optic nerves on OCTA were significantly decreased in patients with CHD after denoising. Retinal VD of the superficial capillary plexus (SCP), deep capillary plexus (DCP) and radial peripapillary capillary (RPC) was strongly correlated with serum BNP levels in patients with CHD. Significant differences were noted in retinal thickness and retinal VD (SCP, DCP and RPC) between the increased BNP and normal BNP groups in patients with CHD. Conclusion Deep learning denoising can remove background noise and smooth rough vessel surfaces. SCP,DCP and RPC may be potential clinical markers of cardiac function in patients with CHD. Denoising shows great potential for improving the sensitivity of OCTA images as a biomarker for CHD progression.
Collapse
Affiliation(s)
- Jin wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Huan Weng
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yiwen Qian
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yuceng Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Luoziyi Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Xin Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Pei Zhang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| | - Zhiliang Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
25
|
Mizoguchi E, Sadanaga T, Nanni L, Wang S, Mizoguchi A. Recently Updated Role of Chitinase 3-like 1 on Various Cell Types as a Major Influencer of Chronic Inflammation. Cells 2024; 13:678. [PMID: 38667293 PMCID: PMC11049018 DOI: 10.3390/cells13080678] [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: 02/27/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Chitinase 3-like 1 (also known as CHI3L1 or YKL-40) is a mammalian chitinase that has no enzymatic activity, but has the ability to bind to chitin, the polymer of N-acetylglucosamine (GlcNAc). Chitin is a component of fungi, crustaceans, arthropods including insects and mites, and parasites, but it is completely absent from mammals, including humans and mice. In general, chitin-containing organisms produce mammalian chitinases, such as CHI3L1, to protect the body from exogenous pathogens as well as hostile environments, and it was thought that it had a similar effect in mammals. However, recent studies have revealed that CHI3L1 plays a pathophysiological role by inducing anti-apoptotic activity in epithelial cells and macrophages. Under chronic inflammatory conditions such as inflammatory bowel disease and chronic obstructive pulmonary disease, many groups already confirmed that the expression of CHI3L1 is significantly induced on the apical side of epithelial cells, and activates many downstream pathways involved in inflammation and carcinogenesis. In this review article, we summarize the expression of CHI3L1 under chronic inflammatory conditions in various disorders and discuss the potential roles of CHI3L1 in those disorders on various cell types.
Collapse
Affiliation(s)
- Emiko Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
- Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI 02912, USA
| | - Takayuki Sadanaga
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
- Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI 02912, USA
| | - Linda Nanni
- Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Siyuan Wang
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
| | - Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
| |
Collapse
|
26
|
Cohen O, Kundel V, Robson P, Al-Taie Z, Suárez-Fariñas M, Shah NA. Achieving Better Understanding of Obstructive Sleep Apnea Treatment Effects on Cardiovascular Disease Outcomes through Machine Learning Approaches: A Narrative Review. J Clin Med 2024; 13:1415. [PMID: 38592223 PMCID: PMC10932326 DOI: 10.3390/jcm13051415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 04/10/2024] Open
Abstract
Obstructive sleep apnea (OSA) affects almost a billion people worldwide and is associated with a myriad of adverse health outcomes. Among the most prevalent and morbid are cardiovascular diseases (CVDs). Nonetheless, randomized controlled trials (RCTs) of OSA treatment have failed to show improvements in CVD outcomes. A major limitation in our field is the lack of precision in defining OSA and specifically subgroups with the potential to benefit from therapy. Further, this has called into question the validity of using the time-honored apnea-hypopnea index as the ultimate defining criteria for OSA. Recent applications of advanced statistical methods and machine learning have brought to light a variety of OSA endotypes and phenotypes. These methods also provide an opportunity to understand the interaction between OSA and comorbid diseases for better CVD risk stratification. Lastly, machine learning and specifically heterogeneous treatment effects modeling can help uncover subgroups with differential outcomes after treatment initiation. In an era of data sharing and big data, these techniques will be at the forefront of OSA research. Advanced data science methods, such as machine-learning analyses and artificial intelligence, will improve our ability to determine the unique influence of OSA on CVD outcomes and ultimately allow us to better determine precision medicine approaches in OSA patients for CVD risk reduction. In this narrative review, we will highlight how team science via machine learning and artificial intelligence applied to existing clinical data, polysomnography, proteomics, and imaging can do just that.
Collapse
Affiliation(s)
- Oren Cohen
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (O.C.); (V.K.)
| | - Vaishnavi Kundel
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (O.C.); (V.K.)
| | - Philip Robson
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Zainab Al-Taie
- Center for Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Z.A.-T.); (M.S.-F.)
| | - Mayte Suárez-Fariñas
- Center for Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (Z.A.-T.); (M.S.-F.)
| | - Neomi A. Shah
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (O.C.); (V.K.)
| |
Collapse
|
27
|
Luo H, Huemer MT, Petrera A, Hauck SM, Rathmann W, Herder C, Koenig W, Hoyer A, Peters A, Thorand B. Association of plasma proteomics with incident coronary heart disease in individuals with and without type 2 diabetes: results from the population-based KORA study. Cardiovasc Diabetol 2024; 23:53. [PMID: 38310303 PMCID: PMC10838466 DOI: 10.1186/s12933-024-02143-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Coronary heart disease (CHD) is a major global health concern, especially among individuals with type 2 diabetes (T2D). Given the crucial role of proteins in various biological processes, this study aimed to elucidate the aetiological role and predictive performance of protein biomarkers on incident CHD in individuals with and without T2D. METHODS The discovery cohort included 1492 participants from the Cooperative Health Research in the Region of Augsburg (KORA) S4 study with 147 incident CHD cases (45 vs. 102 cases in the group with T2D and without T2D, respectively) during 15.6 years of follow-up. The validation cohort included 888 participants from the KORA-Age1 study with 70 incident CHD cases (19 vs. 51 cases in the group with T2D and without T2D, respectively) during 6.9 years of follow-up. We measured 233 plasma proteins related to cardiovascular disease and inflammation using proximity extension assay technology. Associations of proteins with incident CHD were assessed using Cox regression and Mendelian randomization (MR) analysis. Predictive models were developed using priority-Lasso and were evaluated on top of Framingham risk score variables using the C-index, category-free net reclassification index (cfNRI), and relative integrated discrimination improvement (IDI). RESULTS We identified two proteins associated with incident CHD in individuals with and 29 in those without baseline T2D, respectively. Six of these proteins are novel candidates for incident CHD. MR suggested a potential causal role for hepatocyte growth factor in CHD development. The developed four-protein-enriched model for individuals with baseline T2D (ΔC-index: 0.017; cfNRI: 0.253; IDI: 0.051) and the 12-protein-enriched model for individuals without baseline T2D (ΔC-index: 0.054; cfNRI: 0.462; IDI: 0.024) consistently improved CHD prediction in the discovery cohort, while in the validation cohort, significant improvements were only observed for selected performance measures (with T2D: cfNRI: 0.633; without T2D: ΔC-index: 0.038; cfNRI: 0.465). CONCLUSIONS This study identified novel protein biomarkers associated with incident CHD in individuals with and without T2D and reaffirmed previously reported protein candidates. These findings enhance our understanding of CHD pathophysiology and provide potential targets for prevention and treatment.
Collapse
Affiliation(s)
- Hong Luo
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Pettenkofer School of Public Health, Munich, Germany
| | - Marie-Theres Huemer
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764, Neuherberg, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine Universität, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine Universität, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität, Düsseldorf, Germany
| | - Wolfgang Koenig
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Annika Hoyer
- Biostatistics and Medical Biometry, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Pettenkofer School of Public Health, Munich, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764, Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Pettenkofer School of Public Health, Munich, Germany.
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany.
| |
Collapse
|
28
|
Chybowska AD, Gadd DA, Cheng Y, Bernabeu E, Campbell A, Walker RM, McIntosh AM, Wrobel N, Murphy L, Welsh P, Sattar N, Price JF, McCartney DL, Evans KL, Marioni RE. Epigenetic Contributions to Clinical Risk Prediction of Cardiovascular Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004265. [PMID: 38288591 PMCID: PMC10876178 DOI: 10.1161/circgen.123.004265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 11/30/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Cardiovascular disease (CVD) is among the leading causes of death worldwide. The discovery of new omics biomarkers could help to improve risk stratification algorithms and expand our understanding of molecular pathways contributing to the disease. Here, ASSIGN-a cardiovascular risk prediction tool recommended for use in Scotland-was examined in tandem with epigenetic and proteomic features in risk prediction models in ≥12 657 participants from the Generation Scotland cohort. METHODS Previously generated DNA methylation-derived epigenetic scores (EpiScores) for 109 protein levels were considered, in addition to both measured levels and an EpiScore for cTnI (cardiac troponin I). The associations between individual protein EpiScores and the CVD risk were examined using Cox regression (ncases≥1274; ncontrols≥11 383) and visualized in a tailored R application. Splitting the cohort into independent training (n=6880) and test (n=3659) subsets, a composite CVD EpiScore was then developed. RESULTS Sixty-five protein EpiScores were associated with incident CVD independently of ASSIGN and the measured concentration of cTnI (P<0.05), over a follow-up of up to 16 years of electronic health record linkage. The most significant EpiScores were for proteins involved in metabolic, immune response, and tissue development/regeneration pathways. A composite CVD EpiScore (based on 45 protein EpiScores) was a significant predictor of CVD risk independent of ASSIGN and the concentration of cTnI (hazard ratio, 1.32; P=3.7×10-3; 0.3% increase in C-statistic). CONCLUSIONS EpiScores for circulating protein levels are associated with CVD risk independent of traditional risk factors and may increase our understanding of the etiology of the disease.
Collapse
Affiliation(s)
- Aleksandra D. Chybowska
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Danni A. Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Yipeng Cheng
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Elena Bernabeu
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Rosie M. Walker
- School of Psychology, University of Exeter, United Kingdom (R.M.W.)
| | - Andrew M. McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital (A.M.M.), The University of Edinburgh, United Kingdom
| | - Nicola Wrobel
- Edinburgh Clinical Research Facility, Western General Hospital (N.W., L.M.), The University of Edinburgh, United Kingdom
| | - Lee Murphy
- Edinburgh Clinical Research Facility, Western General Hospital (N.W., L.M.), The University of Edinburgh, United Kingdom
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (P.W., N.S.)
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (P.W., N.S.)
| | - Jackie F. Price
- Usher Institute, Old Medical School (J.F.P.), The University of Edinburgh, United Kingdom
| | - Daniel L. McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer (A.D.C., D.A.G., Y.C., E.B., A.C., D.L.M., K.L.E., R.E.M.), The University of Edinburgh, United Kingdom
| |
Collapse
|
29
|
Kuku KO, Oyetoro R, Hashemian M, Livinski AA, Shearer JJ, Joo J, Psaty BM, Levy D, Ganz P, Roger VL. Proteomics for heart failure risk stratification: a systematic review. BMC Med 2024; 22:34. [PMID: 38273315 PMCID: PMC10809595 DOI: 10.1186/s12916-024-03249-7] [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: 09/01/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a complex clinical syndrome with persistently high mortality. High-throughput proteomic technologies offer new opportunities to improve HF risk stratification, but their contribution remains to be clearly defined. We aimed to systematically review prognostic studies using high-throughput proteomics to identify protein signatures associated with HF mortality. METHODS We searched four databases and two clinical trial registries for articles published from 2012 to 2023. HF proteomics studies measuring high numbers of proteins using aptamer or antibody-based affinity platforms on human plasma or serum with outcomes of all-cause or cardiovascular death were included. Two reviewers independently screened articles, extracted data, and assessed the risk of bias. A third reviewer resolved conflicts. We assessed the risk of bias using the Risk Of Bias In Non-randomized Studies-of Exposure tool. RESULTS Out of 5131 unique articles identified, nine articles were included in the review. The nine studies were observational; three used the aptamer platform, and six used the antibody platform. We found considerable heterogeneity across studies in measurement panels, HF definitions, ejection fraction categorization, follow-up duration, and outcome definitions, and a lack of risk estimates for most protein associations. Hence, we proceeded with a systematic review rather than a meta-analysis. In two comparable aptamer studies in patients with HF with reduced ejection fraction, 21 proteins were identified in common for the association with all-cause death. Among these, one protein, WAP four-disulfide core domain protein 2 was also reported in an antibody study on HFrEF and for the association with CV death. We proposed standardized reporting criteria to facilitate the interpretation of future studies. CONCLUSIONS In this systematic review of nine studies evaluating the association of proteomics with mortality in HF, we identified a limited number of proteins common across several studies. Heterogeneity across studies compromised drawing broad inferences, underscoring the importance of standardized approaches to reporting.
Collapse
Affiliation(s)
- Kayode O Kuku
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca Oyetoro
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maryam Hashemian
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alicia A Livinski
- Office of Research Services, Office of the Director, National Institutes of Health Library, National Institutes of Health, Bethesda, MD, USA
| | - Joseph J Shearer
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jungnam Joo
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Daniel Levy
- Laboratory for Cardiovascular Epidemiology and Genomics, Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, USA
| | - Véronique L Roger
- Heart Disease Phenomics Laboratory, Epidemiology and Community Health Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Ljunggren M, Zhou X, Theorell-Haglöw J, Janson C, Franklin KA, Emilsson Ö, Lindberg E. Sleep Apnea Indices Associated with Markers of Inflammation and Cardiovascular Disease: A Proteomic Study in the MUSTACHE Cohort. Ann Am Thorac Soc 2024; 21:165-169. [PMID: 37788298 PMCID: PMC10867909 DOI: 10.1513/annalsats.202305-472rl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/03/2023] [Indexed: 10/05/2023] Open
|
31
|
Olszowka M, Hagström E, Hadziosmanovic N, Ljunggren M, Denchev S, Manolis A, Wallentin L, White HD, Stewart RAH, Held C. Excessive daytime sleepiness, morning tiredness, and prognostic biomarkers in patients with chronic coronary syndrome. Int J Cardiol 2024; 394:131395. [PMID: 37748524 DOI: 10.1016/j.ijcard.2023.131395] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Sleep-related breathing disorders (SRBD) are related to cardiovascular outcomes in patients with chronic coronary syndrome (CCS). Whether SRBD-related symptoms are associated with prognostic biomarkers in patients with CCS is not established. METHODS Associations between frequency (never/rarely, sometimes, often, always) of self-reported SRBD-related symptoms (excessive daytime sleepiness [EDS]; morning tiredness [MT]; loud snoring; multiple awakenings/night; gasping, choking, or apnea when asleep) and levels of biomarkers related to cardiovascular prognosis (high-sensitivity C-reactive protein [hs-CRP], interleukin 6 [IL-6], high-sensitivity cardiac troponin T [hs-cTnT], N-terminal pro B-type natriuretic peptide [NT-proBNP], cystatin C, growth differentiation factor 15 [GDF-15] and lipoprotein-associated phospholipase A2 activity) were assessed at baseline in 15,640 patients with CCS on optimal secondary preventive therapy in the STABILITY trial. Cross-sectional associations were assessed by adjusted linear regression models testing for trends with the never/rarely category serving as reference. RESULTS EDS was associated (geometric mean ratio, 95% confidence interval) with increased levels of IL-6 (often 1.07 [1.03-1.10], always 1.15 [1.10-1.21]), GDF-15 (often 1.03 [1.01-1.06], always 1.07 [1.03-1.11]), NT-proBNP (always 1.22 [1.12-1.33]), and hs-cTnT (always 1.07 [1.01-1.12]). MT was associated with increased levels of IL-6 (often 1.05 [1.01-1.09], always 1.09 [1.04-1.15]), and GDF-15 (always 1.06 [1.03-1.10]). All symptoms were to some degree associated with higher levels of hs-CRP and loud snoring was also associated with decreased levels of NT-proBNP and hs-cTnT. CONCLUSIONS In patients with CCS, stepwise increased frequency of SRBD-related symptoms, such as EDS and MT, were associated with gradually higher levels of IL-6 and GDF-15, each reflecting distinct pathophysiological pathways.
Collapse
Affiliation(s)
- Maciej Olszowka
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden.
| | - Emil Hagström
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | | | - Mirjam Ljunggren
- Department of Medical Sciences, Respiratory-, allergy- and sleep research, Uppsala University, Uppsala, Sweden
| | - Stefan Denchev
- Medical Institute of Ministry of Interior, Sofia, Bulgaria
| | | | - Lars Wallentin
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Ralph A H Stewart
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Claes Held
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Chen QS, Bergman O, Ziegler L, Baldassarre D, Veglia F, Tremoli E, Strawbridge RJ, Gallo A, Pirro M, Smit AJ, Kurl S, Savonen K, Lind L, Eriksson P, Gigante B. A machine learning based approach to identify carotid subclinical atherosclerosis endotypes. Cardiovasc Res 2023; 119:2594-2606. [PMID: 37475157 PMCID: PMC10730242 DOI: 10.1093/cvr/cvad106] [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: 10/07/2022] [Revised: 03/12/2023] [Accepted: 05/05/2023] [Indexed: 07/22/2023] Open
Abstract
AIMS To define endotypes of carotid subclinical atherosclerosis. METHODS AND RESULTS We integrated demographic, clinical, and molecular data (n = 124) with ultrasonographic carotid measurements from study participants in the IMPROVE cohort (n = 3340). We applied a neural network algorithm and hierarchical clustering to identify carotid atherosclerosis endotypes. A measure of carotid subclinical atherosclerosis, the c-IMTmean-max, was used to extract atherosclerosis-related features and SHapley Additive exPlanations (SHAP) to reveal endotypes. The association of endotypes with carotid ultrasonographic measurements at baseline, after 30 months, and with the 3-year atherosclerotic cardiovascular disease (ASCVD) risk was estimated by linear (β, SE) and Cox [hazard ratio (HR), 95% confidence interval (CI)] regression models. Crude estimates were adjusted by common cardiovascular risk factors, and baseline ultrasonographic measures. Improvement in ASCVD risk prediction was evaluated by C-statistic and by net reclassification improvement with reference to SCORE2, c-IMTmean-max, and presence of carotid plaques. An ensemble stacking model was used to predict endotypes in an independent validation cohort, the PIVUS (n = 1061). We identified four endotypes able to differentiate carotid atherosclerosis risk profiles from mild (endotype 1) to severe (endotype 4). SHAP identified endotype-shared variables (age, biological sex, and systolic blood pressure) and endotype-specific biomarkers. In the IMPROVE, as compared to endotype 1, endotype 4 associated with the thickest c-IMT at baseline (β, SE) 0.36 (0.014), the highest number of plaques 1.65 (0.075), the fastest c-IMT progression 0.06 (0.013), and the highest ASCVD risk (HR, 95% CI) (1.95, 1.18-3.23). Baseline and progression measures of carotid subclinical atherosclerosis and ASCVD risk were associated with the predicted endotypes in the PIVUS. Endotypes consistently improved measures of ASCVD risk discrimination and reclassification in both study populations. CONCLUSIONS We report four replicable subclinical carotid atherosclerosis-endotypes associated with progression of atherosclerosis and ASCVD risk in two independent populations. Our approach based on endotypes can be applied for precision medicine in ASCVD prevention.
Collapse
Affiliation(s)
- Qiao Sen Chen
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solnavägen 30, 171 64 Stockholm, Sweden
| | - Otto Bergman
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solnavägen 30, 171 64 Stockholm, Sweden
| | - Louise Ziegler
- Division of Medicine and Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Entrevägen 2, 182 88 Stockholm, Sweden
| | - Damiano Baldassarre
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, Via Vanvitelli 32, 20133 Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
| | - Fabrizio Veglia
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera 1, 48033 Cotignola (RA), Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera 1, 48033 Cotignola (RA), Italy
| | - Rona J Strawbridge
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solnavägen 30, 171 64 Stockholm, Sweden
- Institute of Health and Wellbeing, University of Glasgow, Clarice Pears Building, 90 Byres Road, Glasgow G12 8TB, UK
- Health Data Research, Clarice Pears Building, 90 Byres Road, Glasgow G12 8TB, UK
| | - Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, Sorbonne Université, INSERM UMR1166, APHP, Hôpital Pitié-Salpètriêre, 47 Boulevard de l´Hopital, 75013 Paris, France
| | - Matteo Pirro
- Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Piazzale Menghini 1, 06129 Perugia, Italy
| | - Andries J Smit
- Department of Medicine, University Medical Center Groningen, Groningen & Isala Clinics Zwolle, Dokter Spanjaardweg 29B, 8025 BT Groningen, the Netherlands
| | - Sudhir Kurl
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Yliopistonranta 1 C, Canthia Building, B Wing, FI-70211 Kuopio, Finland
| | - Kai Savonen
- Kuopio Research Institute of Exercise Medicine, Haapaniementie 16, FI-70100 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Science Service Center, Kuopio University Hospital, Yliopsistonranta 1F, FI-70211 Kuopio, Finland
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala Science Park, Dag Hammarskjöldsv 10B, 752 37 Uppsala, Sweden
| | - Per Eriksson
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solnavägen 30, 171 64 Stockholm, Sweden
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solnavägen 30, 171 64 Stockholm, Sweden
- Department of Cardiology, Danderyd University Hospital, Entrevägen 2, 182 88 Stockholm, Sweden
| |
Collapse
|
33
|
Zhang F, Li W, Zhang Y, Wang D, Li J, Li C, He L. lncRNA TPRG1-AS1 Screened the Onset of Acute Coronary Syndromes and Predicted Severity and the Occurrence of MACE During Patients' Hospitalization. J Inflamm Res 2023; 16:5385-5391. [PMID: 38026258 PMCID: PMC10661923 DOI: 10.2147/jir.s435945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Acute coronary syndrome (ACS) is a common acute myocardial ischemia syndrome and is one of the death-related causes of cardiovascular diseases. Identifying biomarkers to indicate disease severity and predict the occurrence of major adverse cardiovascular events (MACE) would benefit the clinical prognosis of ACS. This study estimated the expression and significance of lncRNA TPRG1-AS1 in the onset and development of ACS, aiming to explore a novel biomarker for the diagnosis and prognosis of ACS. Patients and Methods A total of 109 ACS patients and 66 patients who received coronary angiography and excluded ACS were enrolled in this study. TPRG1-AS1 in the serum of study subjects was analyzed by PCR. The significance of TPRG1-AS1 in screening ACS was evaluated by ROC analysis. The association of TPRG1-AS1 with the disease severity of ACS was assessed by Pearson correlation analysis with patients' clinicopathological features. The potential of TPRG1-AS1 in predicting the occurrence of MACE was assessed by logistic regression analysis. Results Significant upregulation of TPRG1-AS1 was observed in ACS patients, which served as a risk factor for ACS and distinguish between ACS patients and the normal group. TPRG1-AS1 was positively correlated with Gensini score, cys-C, cTnI, and NT-proBNP levels of ACS patients, which indicate severe development of ACS. Additionally, increasing serum TPRG1-AS1 was associated with the high incidence of MACE during patients' hospitalization and was identified as a risk factor for MACE in ACS patients. Conclusion Upregulated TPRG1-AS1 in ACS served as a diagnostic biomarker and predicted the severe development of patients.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Wei Li
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Yingying Zhang
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Dong Wang
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Jing Li
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Chengpeng Li
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| | - Liqun He
- Department of Cardiology, Intervention Cardiology Center, Wuhan No.1 Hospital, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
34
|
Siegbahn A, Eriksson N, Assarsson E, Lundberg M, Ballagi A, Held C, Stewart RAH, White HD, Åberg M, Wallentin L. Development and validation of a quantitative Proximity Extension Assay instrument with 21 proteins associated with cardiovascular risk (CVD-21). PLoS One 2023; 18:e0293465. [PMID: 37963145 PMCID: PMC10645335 DOI: 10.1371/journal.pone.0293465] [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: 05/10/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Treatment of cardiovascular diseases (CVD) is a substantial burden to healthcare systems worldwide. New tools are needed to improve precision of treatment by optimizing the balance between efficacy, safety, and cost. We developed a high-throughput multi-marker decision support instrument which simultaneously quantifies proteins associated with CVD. METHODS AND FINDINGS Candidate proteins independently associated with different clinical outcomes were selected from clinical studies by the screening of 368 circulating biomarkers. We then custom-designed a quantitative PEA-panel with 21 proteins (CVD-21) by including recombinant antigens as calibrator samples for normalization and absolute quantification of the proteins. The utility of the CVD-21 tool was evaluated in plasma samples from a case-control cohort of 4224 patients with chronic coronary syndrome (CCS) using multivariable Cox regression analyses and machine learning techniques. The assays in the CVD-21 tool gave good precision and high sensitivity with lower level of determination (LOD) between 0.03-0.7 pg/ml for five of the biomarkers. The dynamic range for the assays was sufficient to accurately quantify the biomarkers in the validation study except for troponin I, which in the modeling was replaced by high-sensitive cardiac troponin T (hs-TnT). We created seven different multimarker models, including a reference model with NT-proBNP, hs-TnT, GDF-15, IL-6, and cystatin C and one model with only clinical variables, for the comparison of the discriminative value of the CVD-21 tool. All models with biomarkers including hs-TnT provided similar discrimination for all outcomes, e.g. c-index between 0.68-0.86 and outperformed models using only clinical variables. Most important prognostic biomarkers were MMP-12, U-PAR, REN, VEGF-D, FGF-23, TFF3, ADM, and SCF. CONCLUSIONS The CVD-21 tool is the very first instrument which with PEA simultaneously quantifies 21 proteins with associations to different CVD. Novel pathophysiologic and prognostic information beyond that of established biomarkers were identified by a number of proteins.
Collapse
Affiliation(s)
- Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | | | | | | | - Claes Held
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Harvey D. White
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Mazidi M, Wright N, Yao P, Kartsonaki C, Millwood IY, Fry H, Said S, Pozarickij A, Pei P, Chen Y, Avery D, Du H, Schmidt DV, Yang L, Lv J, Yu C, Chen J, Hill M, Holmes MV, Howson JMM, Peto R, Collins R, Bennett DA, Walters RG, Li L, Clarke R, Chen Z. Plasma Proteomics to Identify Drug Targets for Ischemic Heart Disease. J Am Coll Cardiol 2023; 82:1906-1920. [PMID: 37940228 PMCID: PMC10641761 DOI: 10.1016/j.jacc.2023.09.804] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Integrated analyses of plasma proteomic and genetic markers in prospective studies can clarify the causal relevance of proteins and discover novel targets for ischemic heart disease (IHD) and other diseases. OBJECTIVES The purpose of this study was to examine associations of proteomics and genetics data with IHD in population studies to discover novel preventive treatments. METHODS We conducted a nested case-cohort study in the China Kadoorie Biobank (CKB) involving 1,971 incident IHD cases and 2,001 subcohort participants who were genotyped and free of prior cardiovascular disease. We measured 1,463 proteins in the stored baseline samples using the OLINK EXPLORE panel. Cox regression yielded adjusted HRs for IHD associated with individual proteins after accounting for multiple testing. Moreover, cis-protein quantitative loci (pQTLs) identified for proteins in genome-wide association studies of CKB and of UK Biobank were used as instrumental variables in separate 2-sample Mendelian randomization (MR) studies involving global CARDIOGRAM+C4D consortium (210,842 IHD cases and 1,378,170 controls). RESULTS Overall 361 proteins were significantly associated at false discovery rate <0.05 with risk of IHD (349 positively, 12 inversely) in CKB, including N-terminal prohormone of brain natriuretic peptide and proprotein convertase subtilisin/kexin type 9. Of these 361 proteins, 212 had cis-pQTLs in CKB, and MR analyses of 198 variants in CARDIOGRAM+C4D identified 13 proteins that showed potentially causal associations with IHD. Independent MR analyses of 307 cis-pQTLs identified in Europeans replicated associations for 4 proteins (FURIN, proteinase-activated receptor-1, Asialoglycoprotein receptor-1, and matrix metalloproteinase-3). Further downstream analyses showed that FURIN, which is highly expressed in endothelial cells, is a potential novel target and matrix metalloproteinase-3 a potential repurposing target for IHD. CONCLUSIONS Integrated analyses of proteomic and genetic data in Chinese and European adults provided causal support for FURIN and multiple other proteins as potential novel drug targets for treatment of IHD.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Hannah Fry
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Saredo Said
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Dan Valle Schmidt
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jun Lv
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Canqing Yu
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Junshi Chen
- China National Center for Food Risk Assessment, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael V Holmes
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | | - Richard Peto
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Liming Li
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China; Key Laboratory of Epidemiology of Major (Peking University), Ministry of Education, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
36
|
Zheng Y, Huang Y, Li H. Hemoglobin albumin lymphocyte and platelet score and all-cause mortality in coronary heart disease: a retrospective cohort study of NHANES database. Front Cardiovasc Med 2023; 10:1241217. [PMID: 38028472 PMCID: PMC10679332 DOI: 10.3389/fcvm.2023.1241217] [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: 06/16/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Aim Anemia, inflammatory status, and malnutrition are all important factors in the prognosis of cardiovascular disease (CVD), and their interactions are also noteworthy. A recent scoring system, the hemoglobin albumin lymphocyte and platelet (HALP) score, combining multi-dimensional metrics, has been used in the prognoses of many diseases except coronary heart disease (CHD). Herein, this study aims to explore the association between HALP score and all-cause mortality in patients with CHD. Methods Demographic and clinical data of adult patients with CHD were extracted from the National Health and Nutrition Examination Surveys (NHANES) database from 2003 to 2018 in this retrospective cohort study. Weighted univariate and multivariate COX proportional hazard models were used for covariates screening and exploration of the association between HALP score and all-cause mortality. The evaluation indexes were hazard ratios (ORs) and 95% confidence intervals (CIs). Kaplan-Meier (KM) curve and the receiver operator characteristic (ROC) curve were used to assess the predictive performance of HALP on CHD prognosis. In addition, subgroup analyses of age and congestive heart failure (CHF) were also performed. Results Among the eligible patients, 657 died of all-cause mortality. After adjusting for the covariates including age, education level, PIR, marital status, smoking, physical activity, total energy intake, CHF, stroke, hypertension, DM, CKD, cancer or malignancy, monocyte, drug for CVD, treatment for anemia, anticoagulants drug, and adrenal cortical steroids, we found that HALP score was negatively associated with the risk of all-cause mortality [HR = 0.83, 95% CI: (0.74-0.93)]. Compared with patients with high HALP scores, those who had lower HALP scores seemed to have a higher risk of all-cause mortality (all P < 0.05). HALP score has a potential predictive value on CHD prognosis with an area under the curve (AUC) of 0.61. Furthermore, in patients aged <65 years, with or without CHF, a lower HALP score was also associated with a higher risk of all-cause mortality (all P < 0.05). Conclusions HALP score has a potential predictive value on CHD prognosis; however, the causal association between HALP score and mortality in patients with CHD needs further exploration.
Collapse
Affiliation(s)
| | | | - Haitao Li
- Department of Cardiology, Hainan Province Clinical Medical Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
37
|
Mahoney SA, Dey AK, Basisty N, Herman AB. Identification and functional analysis of senescent cells in the cardiovascular system using omics approaches. Am J Physiol Heart Circ Physiol 2023; 325:H1039-H1058. [PMID: 37656130 PMCID: PMC10908411 DOI: 10.1152/ajpheart.00352.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, and senescent cells have emerged as key contributors to its pathogenesis. Senescent cells exhibit cell cycle arrest and secrete a range of proinflammatory factors, termed the senescence-associated secretory phenotype (SASP), which promotes tissue dysfunction and exacerbates CVD progression. Omics technologies, specifically transcriptomics and proteomics, offer powerful tools to uncover and define the molecular signatures of senescent cells in cardiovascular tissue. By analyzing the comprehensive molecular profiles of senescent cells, omics approaches can identify specific genetic alterations, gene expression patterns, protein abundances, and metabolite levels associated with senescence in CVD. These omics-based discoveries provide insights into the mechanisms underlying senescence-induced cardiovascular damage, facilitating the development of novel diagnostic biomarkers and therapeutic targets. Furthermore, integration of multiple omics data sets enables a systems-level understanding of senescence in CVD, paving the way for precision medicine approaches to prevent or treat cardiovascular aging and its associated complications.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, Colorado, United States
| | - Amit K Dey
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Nathan Basisty
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Allison B Herman
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| |
Collapse
|
38
|
Ferreira MB, Kobayashi M, Costa RQ, Fonseca T, Brandão M, Oliveira JC, Marinho A, Cyrne Carvalho H, Rodrigues P, Zannad F, Rossignol P, Barros AS, Ferreira JP. Unsupervised clustering to differentiate rheumatoid arthritis patients based on proteomic signatures. Scand J Rheumatol 2023; 52:619-626. [PMID: 37083270 DOI: 10.1080/03009742.2023.2196781] [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: 11/11/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023]
Abstract
OBJECTIVE Patients with rheumatoid arthritis (RA) have different presentations and prognoses. Cluster analysis based on proteomic signatures creates independent phenogroups of patients with different pathophysiological backgrounds. We aimed to identify distinct pathophysiological clusters of RA patients based on circulating proteomic biomarkers. METHOD This was a cohort study including 399 RA patients. Clustering was performed on 94 circulating proteins (92 CVDII Olink®, high-sensitivity troponin T, and C-reactive protein). Unsupervised clustering was performed using a partitioning cluster algorithm. RESULTS The clustering algorithm identified two distinct clusters: cluster 1 (n = 223) and cluster 2 (n = 176). Compared with cluster 1, cluster 2 included older patients with a higher burden of comorbidities (cardiovascular and RA related), more erosive and longer RA duration, more dyspnoea and fatigue, walking a shorter distance in the Six-Minute Walk Test, with more severe diastolic dysfunction, and a 4.5-fold higher risk of death or hospitalization for cardiovascular reasons. Tumour necrosis factor (TNF) receptor superfamily-related pathways were mainly responsible for the model's discriminative ability. CONCLUSION Using unsupervised cluster analysis based on proteomic phenotypes, we identified two clusters of RA patients with distinct biomarkers profiles, clinical characteristics, and different outcomes that could reflect different pathophysiological backgrounds. TNF receptor superfamily-related proteins may be used to distinguish subgroups.
Collapse
Affiliation(s)
- M B Ferreira
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Internal Medicine Department, Hospital da Luz Arrábida, Porto, Portugal
| | - M Kobayashi
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - R Q Costa
- Internal Medicine Department, Centro Hospitalar de Entre o Douro e Vouga, Aveiro, Portugal
| | - T Fonseca
- Internal Medicine Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Unidade de Imunologia Clínica, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - M Brandão
- Unidade de Imunologia Clínica, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - J C Oliveira
- Clinical Chemistry Service, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - A Marinho
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Unidade de Imunologia Clínica, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - H Cyrne Carvalho
- UMIB - Unidade Multidisciplinar de Investigação Biomédica, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Cardiology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - P Rodrigues
- Cardiology Department, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - F Zannad
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - P Rossignol
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - A S Barros
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Heart Failure Clinic, Centro Hospitalar de Vila Nova de Gaia/Espinho, Portugal
| | - J P Ferreira
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Heart Failure Clinic, Centro Hospitalar de Vila Nova de Gaia/Espinho, Portugal
| |
Collapse
|
39
|
Song M, Zhang G, Shi H, Zhu E, Deng L, Shen H. Serum YKL-40 in coronary heart disease: linkage with inflammatory cytokines, artery stenosis, and optimal cut-off value for estimating major adverse cardiovascular events. Front Cardiovasc Med 2023; 10:1242339. [PMID: 38028459 PMCID: PMC10644235 DOI: 10.3389/fcvm.2023.1242339] [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: 06/19/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Objective YKL-40, previously known as chitinase-3-like protein 1 (CHI3L1), is an inflammation-related glycoprotein that promotes atherosclerosis, but its application and optimal cut-off value as a prognostic biomarker in coronary heart disease (CHD) require more clinical evidence. Thus, this prospective study aimed to evaluate the linkage of serum YKL-40 with disease features, inflammatory cytokines, and major adverse cardiovascular events (MACEs) in CHD patients. Methods A total of 410 CHD patients were enrolled for serum YKL-40 determination via enzyme-linked immunosorbent assay. Meanwhile, serum YKL-40 levels in 100 healthy controls (HCs) were also quantified. Results YKL-40 level was higher in CHD patients compared with that in HCs (P < 0.001). YKL-40 was positively linked with hyperlipidemia (P = 0.014), diabetes mellitus (P = 0.001), fasting blood glucose (P = 0.045), C-reactive protein (P < 0.001), the Gensini score (P < 0.001), and stenosis degree (graded by the Gensini score) (P < 0.001) in CHD patients. In addition, an elevated YKL-40 level was associated with increased levels of tumor necrosis factor alpha (P = 0.001), interleukin (IL)-1β (P = 0.001), IL-6 (P < 0.001), and IL-17A (P = 0.002) in CHD patients. The 1-/2-/3-year cumulative MACE rates of CHD patients were 5.5%, 14.4%, and 25.0%, respectively. Regarding the prognostic capability, YKL-40 ≥100 ng/ml (the median cut-off value) (P = 0.003) and YKL-40 ≥150 ng/ml (the third interquartile cut-off value) (P = 0.021) reflected an elevated accumulating MACE rate, whereas accumulating MACE was not different between CHD patients with YKL-40 ≥80 and <80 ng/ml (the first interquartile cut-off value) (P = 0.083). Conclusion Serum YKL-40 is positively linked with inflammatory cytokines and the Gensini score, whose high expression cut-off by 100 and 150 ng/ml estimates a higher MACE risk in CHD patients.
Collapse
Affiliation(s)
- Mowei Song
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guofu Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Erjun Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Li Deng
- Department of Extracorporeal Life Support, The People’s Hospital of Gaozhou, Gaozhou, China
| | - Hongtao Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Mone P, Tesorio T, De Donato A, Cioppa A, Jankauskas SS, Salemme L, Santulli G. A novel urinary proteomic classifier predicts the risk of coronary artery disease. Eur J Prev Cardiol 2023; 30:1535-1536. [PMID: 37075225 PMCID: PMC10562135 DOI: 10.1093/eurjpc/zwad123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461 New York City, NY 10461, USA
- University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | | | | | - Angelo Cioppa
- ‘Montevergine’ Clinic, Mercogliano (Avellino), Italy
| | - Stanislovas S Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461 New York City, NY 10461, USA
- University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | - Luigi Salemme
- ‘Montevergine’ Clinic, Mercogliano (Avellino), Italy
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461 New York City, NY 10461, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461 New York City, NY 10461, USA
| |
Collapse
|
41
|
Srialluri N, Surapaneni A, Schlosser P, Chen TK, Schmidt IM, Rhee EP, Coresh J, Grams ME. Circulating Proteins and Mortality in CKD: A Proteomics Study of the AASK and ARIC Cohorts. Kidney Med 2023; 5:100714. [PMID: 37711886 PMCID: PMC10498294 DOI: 10.1016/j.xkme.2023.100714] [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] [Indexed: 09/16/2023] Open
Abstract
Rationale & Objective Proteomics could provide pathophysiologic insight into the increased risk of mortality in patients with chronic kidney disease (CKD). This study aimed to investigate associations between the circulating proteome and all-cause mortality among patients with CKD. Study Design Observational cohort study. Setting & Participants Primary analysis in 703 participants in the African American Study of Kidney Disease and Hypertension (AASK) and validation in 1,628 participants with CKD in the Atherosclerosis Risk in Communities (ARIC) study who attended visit 5. Exposure Circulating proteins. Outcome All-cause mortality. Analytical Approach Among AASK participants, we evaluated the associations of 6,790 circulating proteins with all-cause mortality using multivariable Cox proportional hazards models. Proteins with significant associations were further studied in ARIC Visit 5 participants with CKD. Results In the AASK cohort, the mean age was 54.5 years, 271 (38.5%) were women, and the mean measured glomerular filtration rate (GFR) was 46 mL/min/1.73 m2. The median follow-up was 9.6 years, and 7 distinct proteins were associated with all-cause mortality at the Bonferroni-level threshold (P < 0.05 of the 6,790) after adjustment for demographics and clinical factors, including baseline measured estimated GFR and proteinuria. In the ARIC visit 5 cohort, the mean age was 77.2 years, 903 (55.5%) were women, the mean estimated GFR was 54 mL/min/1.73 m2 and median follow-up was 6.9 years. Of the 7 proteins found in AASK, 3 (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide) were available in the ARIC data, with all 3 significantly associated with death in ARIC. Limitations Possibility of unmeasured confounding. Cause of death was not known. Conclusions Using large-scale proteomic analysis, proteins were reproducibly associated with mortality in 2 cohorts of participants with CKD. Plain-Language Summary Patients with chronic kidney disease (CKD) have a high risk of premature death, with various pathophysiological processes contributing to this increased risk of mortality. This observational cohort study aimed to investigate the associations between circulating proteins and all-cause mortality in patients with CKD using large-scale proteomic analysis. The study analyzed data from the African American Study of Kidney Disease and Hypertension (AASK) study and validated the findings in the Atherosclerosis Risk in Communities (ARIC) Study. A total of 6,790 circulating proteins were evaluated in AASK, and 7 proteins were significantly associated with all-cause mortality. Three of these proteins (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide (BNP)) were also measured in ARIC and were significantly associated with death. Additional studies assessing biomarkers associated with mortality among patients with CKD are needed to evaluate their use in clinical practice.
Collapse
Affiliation(s)
- Nityasree Srialluri
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Aditya Surapaneni
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| | - Pascal Schlosser
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Teresa K. Chen
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Kidney Health Research Collaborative; Division of Nephrology, Department of Medicine, University of California San Francisco and San Francisco VA Health Care System, San Francisco, California
| | - Insa M. Schmidt
- Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Josef Coresh
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Morgan E. Grams
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| |
Collapse
|
42
|
Wang SF, Wu TT, Zheng YY, Hou XG, Yang HT, Yang Y, Xie X. Serum Globulin to Albumin Ratio as a Novel Predictor of Adverse Clinical Outcomes in Coronary Artery Disease Patients Who Underwent PCI. Rev Cardiovasc Med 2023; 24:278. [PMID: 39077558 PMCID: PMC11273180 DOI: 10.31083/j.rcm2410278] [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: 11/30/2022] [Revised: 04/03/2023] [Accepted: 04/23/2023] [Indexed: 07/31/2024] Open
Abstract
Background Coronary heart disease is one of the main causes of Mortality. Many biological indicators have been used to predict the prognosis of patients with coronary heart disease. The ratio of serum globulin to albumin (GAR) has been used to predict the prognosis of patients with various cancers. It has been proven that GAR is related to the prognosis of patients with stroke. However, GAR's role in cardiovascular disease remains unclear. Our purpose was to investigate the predictive value of GAR on clinical outcomes in post-percutaneous coronary intervention (PCI) patients with coronary artery disease (CAD). Methods From Dec. 2016 to Oct. 2021, a total of 14,994 patients undergoing PCI patients admitted to the First Affiliated Hospital of Xinjiang Medical University were divided into high GAR group (GAR ≥ 0.76, n = 4087) and low GAR group (GAR < 0.76, n = 10,907). The incidence of adverse outcomes including all-cause mortality (ACM), cardiovascular mortality (CM), major adverse cardiovascular events (MACE) and major adverse cardiovascular and cerebrovascular events (MACCE) was compared between the two groups. Multivariate Cox regression was used to adjust for the effects of confounding factors, while hazard ratios (HRs) and 95% confidence intervals (95% CI) were calculated. Median follow-up time was 24 months. Results Compared with the low GAR group, the high GAR group had significantly higher incidence of ACM (6.5% vs. 1.7%, p < 0.001); CM (4.9% vs. 1.2%, p < 0.001), MACE (10.5% vs. 6.7%, p < 0.001), and MACCE (11.3% vs. 7.5%, p < 0.001). Cox regression analysis showed the patients in the high GAR group had a 1.62-fold increased risk for ACM (HR = 2.622, 95% CI: 2.130-3.228, p < 0.01), a 1.782-fold increased risk for CM (HR = 2.782, 95% CI: 2.180-3.550, p < 0.01). There was a 37.2% increased risk for MACE (HR = 1.372, 95% CI: 1.204-1.564, p < 0.01), and 32.4% increased risk for MACCE (HR = 1.324, 95% CI: 1.169-1.500, p < 0.01), compared to the patients in the low GAR group. Conclusions The present study suggested that post-PCI CAD patients with higher GAR presented significantly increased mortality and adverse events GAR level at admission may 296 be considered as part of risk stratification when PCI is possible in patients with coronary heart disease. Clinical Trial Registration The detailed information of the PRACTICE study has been registered on http://Clinicaltrials.gov (Identifier: NCT05174143).
Collapse
Affiliation(s)
- Si-Fan Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830054 Urumqi, Xinjiang, China
| |
Collapse
|
43
|
Ning Y, Wang KY, Min X, Hou XG, Wu TT, Ma YT, Xie X. Cystatin C to Left Ventricular Ejection Fraction Ratio as a Novel Predictor of Adverse Outcomes in Patients with Coronary Artery Disease: A Prospective Cohort Study. Rev Cardiovasc Med 2023; 24:260. [PMID: 39076386 PMCID: PMC11270069 DOI: 10.31083/j.rcm2409260] [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: 03/05/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 07/31/2024] Open
Abstract
Background While both cystatin C and left ventricular ejection fraction (LVEF) revealed established prognostic efficacy in coronary artery disease (CAD), the relationship between cystatin C/left ventricular ejection fraction ratio (CLR) and adverse clinical outcomes among patients with CAD following percutaneous coronary intervention (PCI) remains obscure, to date. Therefore, we sought to assess the predictive efficacy of CLR among CAD patients who underwent PCI in current study. Methods A total of 14,733 participants, including 8622 patients with acute coronary syndrome (ACS) and 6111 patients with stable coronary artery disease (SCAD), were enrolled from a prospective cohort of 15,250 CAD patients who underwent PCI and were admitted to the First Affiliated Hospital of Xinjiang Medical University from 2016 to 2021. The primary outcome of this study was mortality, including all-cause mortality (ACM) and cardiac mortality (CM). The secondary outcomes were major adverse cardiovascular events (MACEs), major adverse cardiac and cerebrovascular events (MACCEs) and nonfatal myocardial infarction (NFMI). For CLR, the optimal cut-off value was determined by utilizing receiver operating characteristic curve analysis (ROC). Subsequently, patients were assigned into two groups: a high-CLR group (CLR ≥ 0.019, n = 3877) and a low-CLR group (CLR < 0.019, n = 10,856), based on optimal cut-off value of 0.019. Lastly, the incidence of outcomes between the two groups was compared. Results The high-CLR group had a higher incidence of ACM (8.8% vs. 0.9%), CM (6.7% vs. 0.6%), MACEs (12.7% vs. 5.9%), MACCEs (13.3% vs. 6.7%), and NFMIs (3.3% vs. 0.9%). After adjusting for confounders, multivariate Cox regression analyses revealed that patients with high-CLR had an 8.163-fold increased risk of ACM (HR = 10.643, 95% CI: 5.525~20.501, p < 0.001), a 10.643-fold increased risk of CM (HR = 10.643, 95% CI: 5.525~20.501, p < 0.001), a 2.352-fold increased risk of MACE (HR = 2.352, 95% CI: 1.754~3.154, p < 0.001), a 2.137-fold increased risk of MACCEs (HR = 2.137, 95% CI: 1.611~2.834, p < 0.001), and a 1.580-fold increased risk of NFMI (HR = 1.580, 95% CI: 1.273~1.960, p < 0.001) compared to patients with low-CLR. Conclusions The current study indicated that a high CLR is a novel and powerful predictor of adverse long-term outcomes in CAD patients who underwent PCI, and that, it is a better predictor for patients wtih SCAD and ACS. Clinical Trial Registration NCT05174143, http://Clinicaltrials.gov.
Collapse
Affiliation(s)
- Yi Ning
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Kai-Yang Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Xuan Min
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical
University, 830011 Urumqi, Xinjiang, China
| |
Collapse
|
44
|
Wereski R, Adamson P, Shek Daud NS, McDermott M, Taggart C, Bularga A, Kimenai DM, Lowry MTH, Tuck C, Anand A, Lowe DJ, Chapman AR, Mills NL. High-Sensitivity Cardiac Troponin for Risk Assessment in Patients With Chronic Coronary Artery Disease. J Am Coll Cardiol 2023; 82:473-485. [PMID: 37532417 DOI: 10.1016/j.jacc.2023.05.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Cardiac troponin is used for risk stratification of patients with acute coronary syndromes; however, the role of testing in other settings remains unclear. OBJECTIVES The aim of this study was to evaluate whether cardiac troponin testing could enhance risk stratification in patients with chronic coronary artery disease independent of disease severity and conventional risk measures. METHODS In a prospective cohort of consecutive patients with symptoms suggestive of stable angina attending for outpatient coronary angiography, high-sensitivity cardiac troponin I was measured before angiography, and clinicians were blinded to the results. The primary outcome was myocardial infarction or cardiovascular death during follow-up. RESULTS In 4,240 patients (age 66 years [IQR: 59-73 years], 33% female), coronary artery disease was identified in 3,888 (92%) who had 255 (6%) primary outcome events during a median follow-up of 2.4 years (IQR: 1.3-3.6 years). In patients with coronary artery disease, troponin concentrations were 2-fold higher in those with an event compared with those without (6.7 ng/L [IQR: 3.2-14.2 ng/L] vs 3.3 ng/L [IQR: 1.7-6.6 ng/L]; P < 0.001). Troponin concentrations were associated with the primary outcome after adjusting for cardiovascular risk factors and coronary artery disease severity (adjusted HR: 2.3; 95% CI: 1.7-3.0, log10 troponin; P < 0.001). A troponin concentration >10 ng/L identified patients with a 50% increase in the risk of myocardial infarction or cardiovascular death. CONCLUSIONS In patients with chronic coronary artery disease, cardiac troponin predicts risk of myocardial infarction or cardiovascular death independent of cardiovascular risk factors and disease severity. Further studies are required to evaluate whether routine testing could inform the selection of high-risk patients for treatment intensification. (Myocardial Injury in Patients Referred for Coronary Angiography [MICA]; ISRCTN15620297).
Collapse
Affiliation(s)
- Ryan Wereski
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom. https://twitter.com/RyanWereski
| | - Philip Adamson
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Nur Shazlin Shek Daud
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael McDermott
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Caelan Taggart
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Anda Bularga
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Dorien M Kimenai
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mathew T H Lowry
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris Tuck
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Atul Anand
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David J Lowe
- University of Glasgow, School of Medicine, Glasgow, United Kingdom
| | - Andrew R Chapman
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom. https://twitter.com/chapdoc1
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
45
|
Wang X, Ren J, Ren H, Song W, Qiao Y, Zhao Y, Linghu L, Cui Y, Zhao Z, Chen L, Qiu L. Diabetes mellitus early warning and factor analysis using ensemble Bayesian networks with SMOTE-ENN and Boruta. Sci Rep 2023; 13:12718. [PMID: 37543637 PMCID: PMC10404250 DOI: 10.1038/s41598-023-40036-5] [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: 11/03/2022] [Accepted: 08/03/2023] [Indexed: 08/07/2023] Open
Abstract
Diabetes mellitus (DM) has become the third chronic non-infectious disease affecting patients after tumor, cardiovascular and cerebrovascular diseases, becoming one of the major public health issues worldwide. Detection of early warning risk factors for DM is key to the prevention of DM, which has been the focus of some previous studies. Therefore, from the perspective of residents' self-management and prevention, this study constructed Bayesian networks (BNs) combining feature screening and multiple resampling techniques for DM monitoring data with a class imbalance in Shanxi Province, China, to detect risk factors in chronic disease monitoring programs and predict the risk of DM. First, univariate analysis and Boruta feature selection algorithm were employed to conduct the preliminary screening of all included risk factors. Then, three resampling techniques, SMOTE, Borderline-SMOTE (BL-SMOTE) and SMOTE-ENN, were adopted to deal with data imbalance. Finally, BNs developed by three algorithms (Tabu, Hill-climbing and MMHC) were constructed using the processed data to find the warning factors that strongly correlate with DM. The results showed that the accuracy of DM classification is significantly improved by the BNs constructed by processed data. In particular, the BNs combined with the SMOTE-ENN resampling improved the most, and the BNs constructed by the Tabu algorithm obtained the best classification performance compared with the hill-climbing and MMHC algorithms. The best-performing joint Boruta-SMOTE-ENN-Tabu model showed that the risk factors of DM included family history, age, central obesity, hyperlipidemia, salt reduction, occupation, heart rate, and BMI.
Collapse
Affiliation(s)
- Xuchun Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiahui Ren
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hao Ren
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenzhu Song
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuchao Qiao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ying Zhao
- Shanxi Centre for Disease Control and Prevention, Taiyuan, 030012, Shanxi, China
| | - Liqin Linghu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Centre for Disease Control and Prevention, Taiyuan, 030012, Shanxi, China
| | - Yu Cui
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhiyang Zhao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Limin Chen
- Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| | - Lixia Qiu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
46
|
Wang M, Gong K, Zhu X, Chen S, Zhou J, Zhang H, Han J, Ma L, Duan Y. Identification of circulating T-cell immunoglobulin and mucin domain 4 as a potential biomarker for coronary heart disease. MedComm (Beijing) 2023; 4:e320. [PMID: 37426678 PMCID: PMC10329472 DOI: 10.1002/mco2.320] [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: 12/09/2022] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, is attenuated in vulnerable plaques of advanced atherosclerosis. T-cell immunoglobulin and mucin domain 4 (TIMD4) is a recognition receptor protein for efferocytosis that has been implicated in atherosclerosis mouse models. However, the role of serum-soluble TIMD4 (sTIMD4) in coronary heart disease (CHD) remains unknown. In this study, we analyzed serum samples collected from two groups: Group 1 (36 healthy controls and 70 CHD patients) and Group 2 (44 chronic coronary syndrome [CCS]) and 81 acute coronary syndrome [ACS] patients). We found that sTIMD4 levels in patients with CHD were significantly higher than those in healthy controls and were also higher in ACS than in CCS patients. The area under the receiver operating characteristic curve was 0.787. Furthermore, our in vitro results showed that low-density lipoprotein/lipopolysaccharide activated p38 mitogen-activated protein kinase, which in turn enhanced a disintegrin and metalloproteinase 17, resulting in increased secretion of sTIMD4. This impairment of macrophage efferocytosis promoted inflammation. Thus, this study is not only the first identification of a potential novel biomarker of CHD, sTIMD4, but also demonstrated its pathogenesis mechanism, providing a new direction for the diagnosis and treatment of CHD.
Collapse
Affiliation(s)
- Mengyao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Xinran Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Shasha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Jie Zhou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Hui Zhang
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
- Key Laboratory of Bioactive Materials of Ministry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
| | - Likun Ma
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yajun Duan
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
47
|
Davidsson P, Eketjäll S, Eriksson N, Walentinsson A, Becker RC, Cavallin A, Bogstedt A, Collén A, Held C, James S, Siegbahn A, Stewart R, Storey RF, White H, Wallentin L. Vascular endothelial growth factor-D plasma levels and VEGFD genetic variants are independently associated with outcomes in patients with cardiovascular disease. Cardiovasc Res 2023; 119:1596-1605. [PMID: 36869765 DOI: 10.1093/cvr/cvad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 03/05/2023] Open
Abstract
AIMS The vascular endothelial growth factor (VEGF) family is involved in pathophysiological mechanisms underlying cardiovascular (CV) diseases. The aim of this study was to investigate the associations between circulating VEGF ligands and/or soluble receptors and CV outcome in patients with acute coronary syndrome (ACS) and chronic coronary syndrome (CCS). METHODS AND RESULTS Levels of VEGF biomarkers, including bFGF, Flt-1, KDR (VEGFR2), PlGF, Tie-2, VEGF-A, VEGF-C, and VEGF-D, were measured in the PLATO ACS cohort (n = 2091, discovery cohort). Subsequently, VEGF-D was also measured in the STABILITY CCS cohort (n = 4015, confirmation cohort) to verify associations with CV outcomes. Associations between plasma VEGF-D and outcomes were analysed by multiple Cox regression models with hazard ratios (HR [95% CI]) comparing the upper vs. the lower quartile of VEGF-D. Genome-wide association study (GWAS) of VEGF-D in PLATO identified SNPs that were used as genetic instruments in Mendelian randomization (MR) meta-analyses vs. clinical endpoints. GWAS and MR were performed in patients with ACS from PLATO (n = 10 013) and FRISC-II (n = 2952), and with CCS from the STABILITY trial (n = 10 786). VEGF-D, KDR, Flt-1, and PlGF showed significant association with CV outcomes. VEGF-D was most strongly associated with CV death (P = 3.73e-05, HR 1.892 [1.419, 2.522]). Genome-wide significant associations with VEGF-D levels were identified at the VEGFD locus on chromosome Xp22. MR analyses of the combined top ranked SNPs (GWAS P-values; rs192812042, P = 5.82e-20; rs234500, P = 1.97e-14) demonstrated a significant effect on CV mortality [P = 0.0257, HR 1.81 (1.07, 3.04) per increase of one unit in log VEGF-D]. CONCLUSION This is the first large-scale cohort study to demonstrate that both VEGF-D plasma levels and VEGFD genetic variants are independently associated with CV outcomes in patients with ACS and CCS. Measurements of VEGF-D levels and/or VEGFD genetic variants may provide incremental prognostic information in patients with ACS and CCS.
Collapse
Affiliation(s)
- Pia Davidsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Susanna Eketjäll
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, 751 85 Uppsala, Sweden
| | - Anna Walentinsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Richard C Becker
- Division of Cardiovascular Health and Disease, Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, 231 Albert Sabin Way ML 0542, Cincinnati, OH, 45267, USA
| | - Anders Cavallin
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Anna Bogstedt
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Anna Collén
- Projects, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Claes Held
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, 751 85 Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Stefan James
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, 751 85 Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, 751 85 Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
- Clinical Chemistry, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | - Ralph Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, 2 Park Road, Grafton, Auckland 1023, New Zealand
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Harvey White
- Green Lane Cardiovascular Service, Auckland City Hospital, 2 Park Road, Grafton, Auckland 1023, New Zealand
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, 751 85 Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| |
Collapse
|
48
|
Kundel V, Cohen O, Khan S, Patel M, Kim-Schulze S, Kovacic J, Suárez-Fariñas M, Shah NA. Advanced Proteomics and Cluster Analysis for Identifying Novel Obstructive Sleep Apnea Subtypes before and after Continuous Positive Airway Pressure Therapy. Ann Am Thorac Soc 2023; 20:1038-1047. [PMID: 36780659 DOI: 10.1513/annalsats.202210-897oc] [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: 10/27/2022] [Accepted: 02/13/2023] [Indexed: 02/15/2023] Open
Abstract
Rationale: Studies have shown elevated inflammatory biomarkers in obstructive sleep apnea (OSA), but data after continuous positive airway pressure (CPAP) treatment are inconsistent. Objectives: We used the Olink proteomics panel to identify unique OSA clusters on the basis of inflammatory protein expression and assess the impact of CPAP therapy. Methods: Adults with newly diagnosed OSA had blood drawn at baseline and three to four months after CPAP. Samples were analyzed using the Olink proteomics platform, which measures 92 prespecified inflammatory proteins using proximity extension assay. Linear mixed-effects models were used to model changes in protein expression during the period of CPAP use, adjusting for batch, age, and sex. Unsupervised hierarchical clustering was performed to identify unique inflammatory OSA clusters on the basis of inflammatory biomarkers. Within-cluster impact of CPAP on inflammatory protein expression was assessed. Results: Among 46 patients, the mean age was 46 ± 12 years (22% women), mean body mass index was 31 ± 5 kg/m2, and mean respiratory disturbance index was 33 ± 17 events/hour. Unsupervised cluster and heatmap analysis revealed three unique proteomic clusters, with low (n = 21), intermediate (n = 19), and high (n = 6) inflammatory protein expression. After CPAP, there were significant within-cluster differences in protein expression. The low inflammatory cluster had a significant increase in protein expression (16%; P = 0.02), and the high inflammatory cluster had a significant decrease in protein expression (-20%; P = 0.003), more significant among those compliant with CPAP in the low (25%; P = 0.04) and high (-22%; P = 0.01) clusters. Conclusions: We identified three unique inflammatory clusters in patients with OSA using plasma proteomics, with a differential response to CPAP by cluster. Our results are hypothesis generating and require further investigation in larger longitudinal studies for enhanced cardiovascular risk profiling in OSA.
Collapse
Affiliation(s)
| | - Oren Cohen
- Division of Pulmonary, Critical Care and Sleep Medicine
| | - Samira Khan
- Division of Pulmonary, Critical Care and Sleep Medicine
| | - Manishkumar Patel
- Human Immune Monitoring Center, Hess Center for Science and Medicine
| | | | - Jason Kovacic
- Cardiovascular Research Institute, and
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; and
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Mayte Suárez-Fariñas
- Center for Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Neomi A Shah
- Division of Pulmonary, Critical Care and Sleep Medicine
| |
Collapse
|
49
|
Dong H, Zhu B, Kong X, Zhang X. Efficient clinical data analysis for prediction of coal workers' pneumoconiosis using machine learning algorithms. THE CLINICAL RESPIRATORY JOURNAL 2023. [PMID: 37380332 PMCID: PMC10363790 DOI: 10.1111/crj.13657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE The purpose of this study is to propose an efficient coal workers' pneumoconiosis (CWP) clinical prediction system and put it into clinical use for clinical diagnosis of pneumoconiosis. METHODS Patients with CWP and dust-exposed workers who were enrolled from August 2021 to December 2021 were included in this study. Firstly, we chose the embedded method through using three feature selection approaches to perform the prediction analysis. Then, we performed the machine learning algorithms as the model backbone and combined them with three feature selection methods, respectively, to determine the optimal predictive model for CWP. RESULTS Through applying three feature selection approaches based on machine learning algorithms, it was found that AaDO2 and some pulmonary function indicators played an important role in prediction for identifying CWP of early stage. The support vector machine (SVM) algorithm was proved as the optimal machine learning model for predicting CWP, with the ROC curves obtained from three feature selection methods using SVM algorithm whose AUC values of 97.78%, 93.7%, and 95.56%, respectively. CONCLUSION We developed the optimal model (SVM algorithm) through comparisons and analyses among the performances of different models for the prediction of CWP as a clinical application.
Collapse
Affiliation(s)
- Hantian Dong
- Department of Geriatric Diseases, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
- National Health Commission Key Laboratory of Pneumoconiosis, Shanxi Province Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Biaokai Zhu
- Network Security Department, Shanxi Police College, Taiyuan, Shanxi, People's Republic of China
| | - Xiaomei Kong
- National Health Commission Key Laboratory of Pneumoconiosis, Shanxi Province Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Xinri Zhang
- National Health Commission Key Laboratory of Pneumoconiosis, Shanxi Province Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
50
|
Sun T, Ye M, Lei F, Qin JJ, Liu YM, Chen Z, Chen MM, Yang C, Zhang P, Ji YX, Zhang XJ, She ZG, Cai J, Jin ZX, Li H. Prevalence and trend of atrial fibrillation and its associated risk factors among the population from nationwide health check-up centers in China, 2012-2017. Front Cardiovasc Med 2023; 10:1151575. [PMID: 37324618 PMCID: PMC10264614 DOI: 10.3389/fcvm.2023.1151575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Background Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, which poses huge disease burdens in China. A study was conducted to systematically analyze the recent prevalence trend of AF and age-related disparities in AF risk among the nationwide healthy check-up population. Method We conducted a nationwide cross-sectional study involving 3,049,178 individuals ≥35 years from health check-up centers to explore the prevalence and trend of AF by age, sex, and region from 2012 to 2017. Additionally, we analyzed risk factors associated with AF among the overall population and different age groups via the Boruta algorithm, the LASSO regression, and the Logistic regression. Result The age-, sex-. and regional-standardized prevalence of AF kept stable between 0.4%-0.45% among national physical examination individuals from 2012 to 2017. However, the prevalence of AF showed an undesirable upward trend in the 35-44-year age group (annual percentage changes (APC): 15.16 [95%CI: 6.42,24.62]). With increasing age, the risk of AF associated with the overweight or obesity gradually exceeds that associated with diabetes and hypertension. In addition to traditional leading risk factors such as age≥65 and coronary heart disease, elevated uric acid and impaired renal function were tightly correlated with AF in the population. Conclusion The significant rise in the prevalence of AF in the 35-44 age group reminds us that in addition to the elderly (the high-risk group), younger people seem to be in more urgent need of attention. Age-related disparities in AF risk also exist. This updated information may provide references for the national prevention and control of AF.
Collapse
Affiliation(s)
- Tao Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
| | - Mao Ye
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Translation Medicine Research Center of Yangtze University, Huanggang, China
| | - Fang Lei
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
| | - Ye-Mao Liu
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Translation Medicine Research Center of Yangtze University, Huanggang, China
| | - Ze Chen
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming-Ming Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
| | - Chengzhang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhao-Xia Jin
- Department of Cardiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Translation Medicine Research Center of Yangtze University, Huanggang, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of ModelAnimal, Wuhan University, Wuhan, China
- Translation Medicine Research Center of Yangtze University, Huanggang, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|