1
|
Nishimura H, Ishii J, Takahashi H, Ishihara Y, Nakamura K, Kitagawa F, Sakaguchi E, Sasaki Y, Kawai H, Muramatsu T, Harada M, Yamada A, Tanizawa-Motoyama S, Naruse H, Sarai M, Yanase M, Ishii H, Watanabe E, Ozaki Y, Izawa H. Prognostic value of combining cardiac myosin-binding protein C and N-terminal pro-B-type natriuretic peptide in patients without acute coronary syndrome treated at medical cardiac intensive care units. Heart Vessels 2025; 40:531-544. [PMID: 39630269 DOI: 10.1007/s00380-024-02492-5] [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: 07/28/2024] [Accepted: 11/20/2024] [Indexed: 01/06/2025]
Abstract
We investigated the prognostic value of cardiac myosin-binding protein C (cMyC), a novel cardiospecific marker, both independently and in combination with N-terminal pro-B-type natriuretic peptide (NT-proBNP), for predicting 6-month all-cause mortality in patients without acute coronary syndrome (ACS) treated at medical (nonsurgical) cardiac intensive care units (CICUs). Admission levels of cMyC, high-sensitivity cardiac troponin T (hs-cTnT), and NT-proBNP were measured in 1032 consecutive patients (mean age; 70 years) without ACS hospitalized acutely in medical CICUs for the treatment of cardiovascular disease. Serum cMyC was closely correlated with hs-cTnT and moderately with NT-proBNP (r = 0.92 and r = 0.49, respectively, p < 0.0001). During the 6-month follow-up period after admission, there were 109 (10.6%) all-cause deaths, including 72 cardiovascular deaths. Both cMyC and NT-proBNP were independent predictors of 6-month all-cause mortality (all p < 0.05). Combining cMyC and NT-proBNP with a baseline model of established risk factors improved patient classification and discrimination beyond any single biomarker (all p < 0.05) or the baseline model alone (both p < 0.0001). Moreover, patients were divided into nine groups using cMyC and NT-proBNP tertiles, and the adjusted hazard ratio (95% confidence interval) for 6-month all-cause mortality in patients with both biomarkers in the highest vs. lowest tertile was 9.67 (2.65-35.2). When cMyC was replaced with hs-cTnT, similar results were observed for hs-cTnT. In addition, the C-indices for addition of cMyC or hs-cTnT to the baseline model were similar (0.798 vs. 0.800, p = 0.94). In conclusion, similar to hs-cTnT, cMyC at admission may be a potent, independent predictor of 6-month all-cause mortality in patients without ACS treated at medical CICUs, and their prognostic abilities may be comparable. Combining cMyC or hs-cTnT with NT-proBNP may substantially improve early risk stratification of this population.
Collapse
Affiliation(s)
- Hideto Nishimura
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Junnichi Ishii
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan.
- Toyota Autobody Yoshiwara Clinic, 25 Kamifujiike, Yoshiwara-cho, Toyota, 473-8517, Japan.
| | - Hiroshi Takahashi
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Yuya Ishihara
- Department of Laboratory of Clinical Medicine, Fujita Health University Hospital, Toyoake, Japan
| | - Kazuhiro Nakamura
- Department of Laboratory of Clinical Medicine, Fujita Health University Hospital, Toyoake, Japan
| | - Fumihiko Kitagawa
- Department of Laboratory of Clinical Medicine, Fujita Health University Hospital, Toyoake, Japan
| | - Eirin Sakaguchi
- Faculty of Medical Technology, School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Yuko Sasaki
- Sysmex R&D Center Europe GmbH, Hamburg, Germany
| | - Hideki Kawai
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Takashi Muramatsu
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Masahide Harada
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Akira Yamada
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Sadako Tanizawa-Motoyama
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Hiroyuki Naruse
- Faculty of Medical Technology, School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Masayoshi Sarai
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Masanobu Yanase
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Hideki Ishii
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Eiichi Watanabe
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| | - Hideo Izawa
- Department of Cardiology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Dengakugakubo, Toyoake, 470-1192, Japan
| |
Collapse
|
2
|
Shahjahan, Dey JK, Dey SK. Translational bioinformatics approach to combat cardiovascular disease and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:221-261. [PMID: 38448136 DOI: 10.1016/bs.apcsb.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Bioinformatics is an interconnected subject of science dealing with diverse fields including biology, chemistry, physics, statistics, mathematics, and computer science as the key fields to answer complicated physiological problems. Key intention of bioinformatics is to store, analyze, organize, and retrieve essential information about genome, proteome, transcriptome, metabolome, as well as organisms to investigate the biological system along with its dynamics, if any. The outcome of bioinformatics depends on the type, quantity, and quality of the raw data provided and the algorithm employed to analyze the same. Despite several approved medicines available, cardiovascular disorders (CVDs) and cancers comprises of the two leading causes of human deaths. Understanding the unknown facts of both these non-communicable disorders is inevitable to discover new pathways, find new drug targets, and eventually newer drugs to combat them successfully. Since, all these goals involve complex investigation and handling of various types of macro- and small- molecules of the human body, bioinformatics plays a key role in such processes. Results from such investigation has direct human application and thus we call this filed as translational bioinformatics. Current book chapter thus deals with diverse scope and applications of this translational bioinformatics to find cure, diagnosis, and understanding the mechanisms of CVDs and cancers. Developing complex yet small or long algorithms to address such problems is very common in translational bioinformatics. Structure-based drug discovery or AI-guided invention of novel antibodies that too with super-high accuracy, speed, and involvement of considerably low amount of investment are some of the astonishing features of the translational bioinformatics and its applications in the fields of CVDs and cancers.
Collapse
Affiliation(s)
- Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
3
|
Hasselbalch RB, Alaour B, Kristensen JH, Couch LS, Kaier TE, Nielsen TL, Plesner LL, Strandkjær N, Schou M, Rydahl C, Goetze JP, Bundgaard H, Marber M, Iversen KK. Hemodialysis and biomarkers of myocardial infarction - a cohort study. Clin Chem Lab Med 2024; 62:361-370. [PMID: 37556843 DOI: 10.1515/cclm-2023-0071] [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: 01/18/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVES End-stage renal disease is associated with a high risk of cardiovascular disease. We compared the concentration and prognostic ability of high sensitivity cardiac troponin T (hs-cTnT) and I (hs-cTnI) and cardiac myosin-binding protein C (cMyC) among stable hemodialysis patients. METHODS Patients were sampled before and after hemodialysis. We measured hs-cTnI, hs-cTnT and cMyC and used Cox regressions to assess the association between quartiles of concentrations and all-cause mortality and a combination of cardiovascular events and all-cause mortality during follow-up. RESULTS A total of 307 patients were included, 204 males, mean age 66 years (SD 14). Before dialysis, 299 (99 %) had a hs-cTnT concentration above the 99th percentile, compared to 188 (66 %) for cMyC and 35 (11 %) for hs-cTnI. Hs-cTnT (23 %, p<0.001) and hs-cTnI (15 %, p=0.049) but not cMyC (4 %, p=0.256) decreased during dialysis. Follow-up was a median of 924 days (492-957 days); patients in the 3rd and 4th quartiles of hs-cTnT (3rd:HR 3.0, 95 % CI 1.5-5.8, 4th:5.2, 2.7-9.8) and the 4th quartile of hs-cTnI (HR 3.8, 2.2-6.8) had an increased risk of mortality. Both were associated with an increased risk of the combined endpoint for patients in the 3rd and 4th quartiles. cMyC concentrations were not associated with risk of mortality or cardiovascular event. CONCLUSIONS Hs-cTnT was above the 99th percentile in almost all patients. This was less frequent for hs-cTnI and cMyC. High cTn levels were associated with a 3-5-fold higher mortality. This association was not present for cMyC. These findings are important for management of hemodialysis patients.
Collapse
Affiliation(s)
- Rasmus Bo Hasselbalch
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Bashir Alaour
- King's College London British Heart Foundation Centre, Rayne Institute, St Thomas' Hospital, London, UK
| | - Jonas Henrik Kristensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Liam S Couch
- King's College London British Heart Foundation Centre, Rayne Institute, St Thomas' Hospital, London, UK
| | - Thomas E Kaier
- King's College London British Heart Foundation Centre, Rayne Institute, St Thomas' Hospital, London, UK
| | - Ture Lange Nielsen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Louis Lind Plesner
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Nina Strandkjær
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Morten Schou
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Casper Rydahl
- Department of Nephrology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Michael Marber
- King's College London British Heart Foundation Centre, Rayne Institute, St Thomas' Hospital, London, UK
| | - Kasper Karmark Iversen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Emergency Medicine, Copenhagen University Hospital - Herlev and Gentofte Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Zhan C, Tang T, Wu E, Zhang Y, He M, Wu R, Bi C, Wang J, Zhang Y, Shen B. From multi-omics approaches to personalized medicine in myocardial infarction. Front Cardiovasc Med 2023; 10:1250340. [PMID: 37965091 PMCID: PMC10642346 DOI: 10.3389/fcvm.2023.1250340] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Myocardial infarction (MI) is a prevalent cardiovascular disease characterized by myocardial necrosis resulting from coronary artery ischemia and hypoxia, which can lead to severe complications such as arrhythmia, cardiac rupture, heart failure, and sudden death. Despite being a research hotspot, the etiological mechanism of MI remains unclear. The emergence and widespread use of omics technologies, including genomics, transcriptomics, proteomics, metabolomics, and other omics, have provided new opportunities for exploring the molecular mechanism of MI and identifying a large number of disease biomarkers. However, a single-omics approach has limitations in understanding the complex biological pathways of diseases. The multi-omics approach can reveal the interaction network among molecules at various levels and overcome the limitations of the single-omics approaches. This review focuses on the omics studies of MI, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, and other omics. The exploration extended into the domain of multi-omics integrative analysis, accompanied by a compilation of diverse online resources, databases, and tools conducive to these investigations. Additionally, we discussed the role and prospects of multi-omics approaches in personalized medicine, highlighting the potential for improving diagnosis, treatment, and prognosis of MI.
Collapse
Affiliation(s)
- Chaoying Zhan
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Tang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Erman Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengqiao He
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rongrong Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Bi
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jiao Wang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yingbo Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Bairong Shen
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Schulte C, Singh B, Theofilatos K, Sörensen NA, Lehmacher J, Hartikainen T, Haller PM, Westermann D, Zeller T, Blankenberg S, Neumann JT, Mayr M. Serial measurements of protein and microRNA biomarkers to specify myocardial infarction subtypes. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 1:None. [PMID: 36185590 PMCID: PMC9514835 DOI: 10.1016/j.jmccpl.2022.100014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/10/2022] [Indexed: 11/19/2022]
Abstract
Background While cardiac-specific troponin (cTn) allows for rapid diagnosis of acute type 1 myocardial infarction (T1MI), its performance to differentiate acute myocardial injury (AI) or type 2 myocardial infarction (T2MI) is limited. The objective was to combine biomarkers to improve discrimination of different myocardial infarction (MI) aetiologies. Methods We determined levels of cardiac troponin T and I (cTnT, cTnI), cardiac myosin-binding protein C (cMyBP-C), NT-proBNP and ten miRNAs, known to be associated with cardiac pathology in a total of n = 495 serial plasma samples at three time points (on admission, after 1 h and 3 h) from 57 NSTEMI (non-ST-elevation myocardial infarction), 18 AI, and 31 STEMI patients, as defined by fourth universal definition of MI (UDMI4) and 59 control individuals. We then applied linear mixed effects model to compare the kinetics of all molecules in these MI sub-types. Results Established (cTnT, cTnI) and novel (cMyBP-C) cardiac necrosis markers failed in differentiating T1MI vs T2MI at early time points. All cardiac necrosis markers were higher in T1MI than in T2MI at 3 h after admission. Muscle-enriched miRNAs (miR-1 and miR-133a) were correlated with cardiac necrosis protein markers and did not offer better discrimination. Established cardiac strain marker NT-proBNP differentiated AI and T1MI at all time points but failed to discriminate T2MI from T1MI. However, the combination of NT-proBNP and cTnT along with age returned an overall AUC of 0.76 [95 % CI 0.67-0.84] for differentiating T1MI, T2MI and AI. Conclusions Rather than using single biomarkers of myocardial necrosis, a combination of clinical biomarkers for cardiac necrosis (troponin) and cardiac strain (NT-proBNP) might aid in differentiating T1MI, T2MI and AI.
Collapse
Affiliation(s)
- Christian Schulte
- King's British Heart Foundation Centre, King's College London, United Kingdom
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Bhawana Singh
- King's British Heart Foundation Centre, King's College London, United Kingdom
| | | | - Nils A. Sörensen
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Jonas Lehmacher
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Tau Hartikainen
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paul M. Haller
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tanja Zeller
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
- University Center of Cardiovascular Science, Hamburg, Germany
| | - Stefan Blankenberg
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Johannes T. Neumann
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, United Kingdom
| |
Collapse
|
6
|
Dar MA, Arafah A, Bhat KA, Khan A, Khan MS, Ali A, Ahmad SM, Rashid SM, Rehman MU. Multiomics technologies: role in disease biomarker discoveries and therapeutics. Brief Funct Genomics 2022; 22:76-96. [PMID: 35809340 DOI: 10.1093/bfgp/elac017] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/21/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Medical research has been revolutionized after the publication of the full human genome. This was the major landmark that paved the way for understanding the biological functions of different macro and micro molecules. With the advent of different high-throughput technologies, biomedical research was further revolutionized. These technologies constitute genomics, transcriptomics, proteomics, metabolomics, etc. Collectively, these high-throughputs are referred to as multi-omics technologies. In the biomedical field, these omics technologies act as efficient and effective tools for disease diagnosis, management, monitoring, treatment and discovery of certain novel disease biomarkers. Genotyping arrays and other transcriptomic studies have helped us to elucidate the gene expression patterns in different biological states, i.e. healthy and diseased states. Further omics technologies such as proteomics and metabolomics have an important role in predicting the role of different biological molecules in an organism. It is because of these high throughput omics technologies that we have been able to fully understand the role of different genes, proteins, metabolites and biological pathways in a diseased condition. To understand a complex biological process, it is important to apply an integrative approach that analyses the multi-omics data in order to highlight the possible interrelationships of the involved biomolecules and their functions. Furthermore, these omics technologies offer an important opportunity to understand the information that underlies disease. In the current review, we will discuss the importance of omics technologies as promising tools to understand the role of different biomolecules in diseases such as cancer, cardiovascular diseases, neurodegenerative diseases and diabetes. SUMMARY POINTS
Collapse
|
7
|
Alaour B, Omland T, Torsvik J, Kaier TE, Sylte MS, Strand H, Quraishi J, McGrath S, Williams L, Meex S, Redwood S, Marber M, Aakre KM. Biological variation of cardiac myosin-binding protein C in healthy individuals. Clin Chem Lab Med 2022; 60:576-583. [PMID: 34162037 DOI: 10.1515/cclm-2021-0306] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/10/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Cardiac myosin-binding protein C (cMyC) is a novel biomarker of myocardial injury, with a promising role in the triage and risk stratification of patients presenting with acute cardiac disease. In this study, we assess the weekly biological variation of cMyC, to examine its potential in monitoring chronic myocardial injury, and to suggest analytical quality specification for routine use of the test in clinical practice. METHODS Thirty healthy volunteers were included. Non-fasting samples were obtained once a week for ten consecutive weeks. Samples were tested in duplicate on the Erenna® platform by EMD Millipore Corporation. Outlying measurements and subjects were identified and excluded systematically, and homogeneity of analytical and within-subject variances was achieved before calculating the biological variability (CVI and CVG), reference change values (RCV) and index of individuality (II). RESULTS Mean age was 38 (range, 21-64) years, and 16 participants were women (53%). The biological variation, RCV and II with 95% confidence interval (CI) were: CVA (%) 19.5 (17.8-21.6), CVI (%) 17.8 (14.8-21.0), CVG (%) 66.9 (50.4-109.9), RCV (%) 106.7 (96.6-120.1)/-51.6 (-54.6 to -49.1) and II 0.42 (0.29-0.56). There was a trend for women to have lower CVG. The calculated RCVs were comparable between genders. CONCLUSIONS cMyC exhibits acceptable RCV and low II suggesting that it could be suitable for disease monitoring, risk stratification and prognostication if measured serially. Analytical quality specifications based on biological variation are similar to those for cardiac troponin and should be achievable at clinically relevant concentrations.
Collapse
Affiliation(s)
- Bashir Alaour
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Janniche Torsvik
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Thomas E Kaier
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Marit S Sylte
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Heidi Strand
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway
| | - Jasmine Quraishi
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | | | | | - Steven Meex
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, The Netherlands
| | - Simon Redwood
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Kristin M Aakre
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Miura T, Kuno A, Tanaka M. Diabetes modulation of the myocardial infarction- acute kidney injury axis. Am J Physiol Heart Circ Physiol 2022; 322:H394-H405. [PMID: 35089809 DOI: 10.1152/ajpheart.00639.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since there is crosstalk in functions of the heart and kidney, acute or chronic injury in one of the two organs provokes adaptive and/or maladaptive responses in both organs, leading to cardiorenal syndrome (CRS). Acute kidney injury (AKI) induced by acute heart failure is referred to as type 1 CRS, and a frequent cause of this type of CRS is acute myocardial infarction (AMI). Diabetes mellitus increases the risk of AMI and also the risk of AKI of various causes. However, there have been only a few studies in which animal models of diabetes were used to examine how diabetes modulates AMI-induced AKI. In this review, we summarize findings regarding the mechanisms of type 1 CRS and the impact of diabetes on both AMI and renal susceptibility to AKI and we discuss mechanisms by which diabetes modulates AMI-induced AKI. Hemodynamic alterations induced by AMI could be augmented by diabetes via its detrimental effect on infarct size and contractile function of the non-infarcted region in the heart. Diabetes increases susceptibility of renal cells to hypoxia and oxidative stress by modulation of signaling pathways that regulate cell survival and autophagy. Recent studies have shown that diabetes mellitus even at early stage of cardiomyopathy/nephropathy predisposes the kidney to AMI-induced AKI, in which activation of toll-like receptors and reactive oxygen species derived from NADPH oxidases are involved. Further analysis of crosstalk between diabetic cardiomyopathy and diabetic kidney disease is necessary for obtaining a more comprehensive understanding of modulation of the AMI-AKI axis by diabetes.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan.,Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
9
|
Wang J, He J, Fan Y, Xu F, Liu Q, He R, Yan R. Extensive mitochondrial proteome disturbance occurs during the early stages of acute myocardial ischemia. Exp Ther Med 2021; 23:85. [PMID: 34938367 PMCID: PMC8688935 DOI: 10.3892/etm.2021.11008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial malfunction leads to the remodeling of myocardial energy metabolism during myocardial ischemia (MI). However, the alterations to the mitochondrial proteome profile during this period has not yet been clarified. An acute MI model was established by high position ligation of the left anterior descending artery in 8-week-old C57BL/6N mice. After 15 min of ligation, the animals were euthanized, and their hearts were collected. The myocardial ultrastructure was observed using transmission electron microscopy (TEM). The cardiac mitochondrial proteome profile was analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and bioinformatics analyses. TEM showed that the outer membrane of the mitochondria was dissolved, and the inner membrane (cristae) was corrupted and broken down extensively in the MI group. The mitochondrial membrane potential was decreased. More than 1,700 mitochondrial proteins were identified by LC-MS/MS analysis, and 119 were differentially expressed. Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis showed that endopeptidase activity regulation, the mitochondrial inner membrane, oxidative phosphorylation, the hypoxia-inducible factor-1 signaling pathway, the pentose phosphate pathway and the peroxisome proliferator-activated receptor signaling pathway were involved in the pathophysiological process in the early stage of acute MI. Extensive and substantial changes in the mitochondrial proteins as well as mitochondrial microstructural damage occur in the early stages of acute MI. In the present study, the series of proteins crucially involved in the pathways of mitochondrial dysfunction and metabolism were identified. Further studies are needed to clarify the roles of these proteins in myocardial metabolism remodeling during acute MI injury.
Collapse
Affiliation(s)
- Jie Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Jun He
- Department of Cardiovascular Internal Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Yucheng Fan
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Fangjing Xu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Qian Liu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Ruhua He
- Department of Cardiovascular Internal Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| | - Ru Yan
- Department of Cardiovascular Internal Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R China
| |
Collapse
|
10
|
Joshi A, Rienks M, Theofilatos K, Mayr M. Systems biology in cardiovascular disease: a multiomics approach. Nat Rev Cardiol 2021; 18:313-330. [PMID: 33340009 DOI: 10.1038/s41569-020-00477-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Omics techniques generate large, multidimensional data that are amenable to analysis by new informatics approaches alongside conventional statistical methods. Systems theories, including network analysis and machine learning, are well placed for analysing these data but must be applied with an understanding of the relevant biological and computational theories. Through applying these techniques to omics data, systems biology addresses the problems posed by the complex organization of biological processes. In this Review, we describe the techniques and sources of omics data, outline network theory, and highlight exemplars of novel approaches that combine gene regulatory and co-expression networks, proteomics, metabolomics, lipidomics and phenomics with informatics techniques to provide new insights into cardiovascular disease. The use of systems approaches will become necessary to integrate data from more than one omic technique. Although understanding the interactions between different omics data requires increasingly complex concepts and methods, we argue that hypothesis-driven investigations and independent validation must still accompany these novel systems biology approaches to realize their full potential.
Collapse
Affiliation(s)
- Abhishek Joshi
- King's British Heart Foundation Centre, King's College London, London, UK
- Bart's Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Marieke Rienks
- King's British Heart Foundation Centre, King's College London, London, UK
| | | | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK.
| |
Collapse
|
11
|
Deguchi H, Morla S, Griffin JH. Novel blood coagulation molecules: Skeletal muscle myosin and cardiac myosin. J Thromb Haemost 2021; 19:7-19. [PMID: 32920971 PMCID: PMC7819347 DOI: 10.1111/jth.15097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Essentials Striated muscle myosins can promote prothrombin activation by FXa or FVa inactivation by APC. Cardiac myosin and skeletal muscle myosin are pro-hemostatic in murine tail cut bleeding models. Infused cardiac myosin exacerbates myocardial injury caused by myocardial ischemia reperfusion. Skeletal muscle myosin isoforms that circulate in human plasma can be grouped into 3 phenotypes. ABSTRACT: Two striated muscle myosins, namely skeletal muscle myosin (SkM) and cardiac myosin (CM), may potentially contribute to physiologic mechanisms for regulation of thrombosis and hemostasis. Thrombin is generated from activation of prothrombin by the prothrombinase (IIase) complex comprising factor Xa, factor Va, and Ca++ ions located on surfaces where these factors are assembled. We discovered that SkM and CM, which are abundant motor proteins in skeletal and cardiac muscles, can provide a surface for thrombin generation by the prothrombinase complex without any apparent requirement for phosphatidylserine or lipids. These myosins can also provide a surface that supports the inactivation of factor Va by activated protein C/protein S, resulting in negative feedback downregulation of thrombin generation. Although the physiologic significance of these reactions remains to be established for humans, substantive insights may be gleaned from murine studies. In mice, exogenously infused SkM and CM can promote hemostasis as they are capable of reducing tail cut bleeding. In a murine myocardial ischemia-reperfusion injury model, exogenously infused CM exacerbates myocardial infarction damage. Studies of human plasmas show that SkM antigen isoforms of different MWs circulate in human plasma, and they can be used to identify three plasma SkM phenotypes. A pilot clinical study showed that one SkM isoform pattern appeared to be linked to isolated pulmonary embolism. These discoveries enable multiple preclinical and clinical studies of SkM and CM, which should provide novel mechanistic insights with potential translational relevance for the roles of CM and SkM in the pathobiology of hemostasis and thrombosis.
Collapse
Affiliation(s)
- Hiroshi Deguchi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Shravan Morla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Division of Hematology, Department of Medicine, University of California, San Diego, CA, USA
| |
Collapse
|
12
|
Shrivastava A, Haase T, Zeller T, Schulte C. Biomarkers for Heart Failure Prognosis: Proteins, Genetic Scores and Non-coding RNAs. Front Cardiovasc Med 2020; 7:601364. [PMID: 33330662 PMCID: PMC7719677 DOI: 10.3389/fcvm.2020.601364] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is a complex disease in which cardiomyocyte injury leads to a cascade of inflammatory and fibrosis pathway activation, thereby causing decrease in cardiac function. As a result, several biomolecules are released which can be identified easily in circulating body fluids. The complex biological processes involved in the development and worsening of HF require an early treatment strategy to stop deterioration of cardiac function. Circulating biomarkers provide not only an ideal platform to detect subclinical changes, their clinical application also offers the opportunity to monitor disease treatment. Many of these biomarkers can be quantified with high sensitivity; allowing their clinical application to be evaluated beyond diagnostic purposes as potential tools for HF prognosis. Though the field of biomarkers is dominated by protein molecules, non-coding RNAs (microRNAs, long non-coding RNAs, and circular RNAs) are novel and promising biomarker candidates that encompass several ideal characteristics required in the biomarker field. The application of genetic biomarkers as genetic risk scores in disease prognosis, albeit in its infancy, holds promise to improve disease risk estimation. Despite the multitude of biomarkers that have been available and identified, the majority of novel biomarker candidates are not cardiac-specific, and instead may simply be a readout of systemic inflammation or other pathological processes. Thus, the true value of novel biomarker candidates in HF prognostication remains unclear. In this article, we discuss the current state of application of protein, genetic as well as non-coding RNA biomarkers in HF risk prognosis.
Collapse
Affiliation(s)
- Apurva Shrivastava
- Clinic for Cardiology, University Heart and Vascular Center, University Medical Center Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, University Medical Center Eppendorf, Hamburg, Germany
| | - Tina Haase
- Clinic for Cardiology, University Heart and Vascular Center, University Medical Center Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, University Medical Center Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- Clinic for Cardiology, University Heart and Vascular Center, University Medical Center Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, University Medical Center Eppendorf, Hamburg, Germany
| | - Christian Schulte
- Clinic for Cardiology, University Heart and Vascular Center, University Medical Center Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, University Medical Center Eppendorf, Hamburg, Germany.,King's British Heart Foundation Centre, King's College London, London, United Kingdom
| |
Collapse
|
13
|
Noncoding RNAs versus Protein Biomarkers in Cardiovascular Disease. Trends Mol Med 2020; 26:583-596. [PMID: 32470385 DOI: 10.1016/j.molmed.2020.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/23/2020] [Accepted: 02/06/2020] [Indexed: 12/31/2022]
Abstract
The development of more sensitive protein biomarker assays results in continuous improvements in detectability, extending the range of clinical applications to the detection of subclinical cardiovascular disease (CVD). However, these efforts have not yet led to improvements in risk assessment compared with existing risk scores. Noncoding RNAs (ncRNAs) have been assessed as biomarkers, and miRNAs have attracted most attention. More recently, other ncRNA classes have been identified, including long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs). Here, we compare emerging ncRNA biomarkers in the cardiovascular field with protein biomarkers for their potential in clinical application, focusing on myocardial injury.
Collapse
|
14
|
Zilberman-Rudenko J, Deguchi H, Shukla M, Oyama Y, Orje JN, Guo Z, Wyseure T, Mosnier LO, McCarty OJT, Ruggeri ZM, Eckle T, Griffin JH. Cardiac Myosin Promotes Thrombin Generation and Coagulation In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2020; 40:901-913. [PMID: 32102568 DOI: 10.1161/atvbaha.120.313990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Cardiac myosin (CM) is structurally similar to skeletal muscle myosin, which has procoagulant activity. Here, we evaluated CM's ex vivo, in vivo, and in vitro activities related to hemostasis and thrombosis. Approach and Results: Perfusion of fresh human blood over CM-coated surfaces caused thrombus formation and fibrin deposition. Addition of CM to blood passing over collagen-coated surfaces enhanced fibrin formation. In a murine ischemia/reperfusion injury model, exogenous CM, when administered intravenously, augmented myocardial infarction and troponin I release. In hemophilia A mice, intravenously administered CM reduced tail-cut-initiated bleeding. These data provide proof of concept for CM's in vivo procoagulant properties. In vitro studies clarified some mechanisms for CM's procoagulant properties. Thrombin generation assays showed that CM, like skeletal muscle myosin, enhanced thrombin generation in human platelet-rich and platelet-poor plasmas and also in mixtures of purified factors Xa, Va, and prothrombin. Binding studies showed that CM, like skeletal muscle myosin, directly binds factor Xa, supporting the concept that the CM surface is a site for prothrombinase assembly. In tPA (tissue-type plasminogen activator)-induced plasma clot lysis assays, CM was antifibrinolytic due to robust CM-dependent thrombin generation that enhanced activation of TAFI (thrombin activatable fibrinolysis inhibitor). CONCLUSIONS CM in vitro is procoagulant and prothrombotic. CM in vivo can augment myocardial damage and can be prohemostatic in the presence of bleeding. CM's procoagulant and antifibrinolytic activities likely involve, at least in part, its ability to bind factor Xa and enhance thrombin generation. Future work is needed to clarify CM's pathophysiology and its mechanistic influences on hemostasis or thrombosis.
Collapse
Affiliation(s)
- Jevgenia Zilberman-Rudenko
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.).,Department of Biomedical Engineering (J.Z.-R., O.J.T.M.), School of Medicine, Oregon Health & Science University, Portland
| | - Hiroshi Deguchi
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Meenal Shukla
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Yoshimasa Oyama
- Department of Hematology-Oncology (O.J.T.M.), School of Medicine, Oregon Health & Science University, Portland
| | - Jennifer N Orje
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Zihan Guo
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Tine Wyseure
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Laurent O Mosnier
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Owen J T McCarty
- Department of Biomedical Engineering (J.Z.-R., O.J.T.M.), School of Medicine, Oregon Health & Science University, Portland
| | - Zaverio M Ruggeri
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.)
| | - Tobias Eckle
- Department of Hematology-Oncology (O.J.T.M.), School of Medicine, Oregon Health & Science University, Portland
| | - John H Griffin
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA (J.Z.-R., H.D., M.S., J.N.O., Z.G., T.W., L.O.M., Z.M.R., J.H.G.).,Department of Anesthesiology, University of Colorado School of Medicine, Aurora (Y.O., T.E.)
| |
Collapse
|
15
|
Schulte C, Zeller T. Biomarkers in primary prevention : Meaningful diagnosis based on biomarker scores? Herz 2019; 45:10-16. [PMID: 31807790 DOI: 10.1007/s00059-019-04874-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cardiovascular (CV) risk assessment is based on the utilization of risk scores, enabling clinicians to estimate an individual's risk to develop CV pathologies and events. Such risk scores comprise classic CV risk factors such as smoking, diabetes, hypertension, and blood cholesterol levels. Recently, other CV biomarkers such as cardiac troponins have been suggested and evaluated as alternative biomarkers not only in the acute diagnostic setting of myocardial infarction, but also as markers for risk stratification in the general population. In this review, we summarize the current knowledge on biomarkers in the field of primary prevention in cardiovascular disease (CVD). Furthermore, we present potential alternative biomarker-based strategies for CV risk assessment. In this respect we provide an outlook on the potential use of genomic variation as well as circulating non-coding RNAs to complement current risk assessment strategies so as to further personalize risk stratification in CVD.
Collapse
Affiliation(s)
- Christian Schulte
- Klinik für Kardiologie, Universitäres Herz- und Gefäßzentrum Hamburg, Martinistraße 52, 20246, Hamburg, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Hamburg/Lübeck/Kiel, Hamburg, Germany
| | - Tanja Zeller
- Klinik für Kardiologie, Universitäres Herz- und Gefäßzentrum Hamburg, Martinistraße 52, 20246, Hamburg, Germany. .,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Standort Hamburg/Lübeck/Kiel, Hamburg, Germany.
| |
Collapse
|
16
|
Schulte C, Barwari T, Joshi A, Theofilatos K, Zampetaki A, Barallobre-Barreiro J, Singh B, Sörensen NA, Neumann JT, Zeller T, Westermann D, Blankenberg S, Marber M, Liebetrau C, Mayr M. Comparative Analysis of Circulating Noncoding RNAs Versus Protein Biomarkers in the Detection of Myocardial Injury. Circ Res 2019; 125:328-340. [PMID: 31159652 PMCID: PMC6641471 DOI: 10.1161/circresaha.119.314937] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/23/2019] [Accepted: 06/03/2019] [Indexed: 01/16/2023]
Abstract
RATIONALE Noncoding RNAs (ncRNAs), including microRNAs (miRNAs), circular RNAs (circRNAs), and long noncoding RNAs (lncRNAs), are proposed novel biomarkers of myocardial injury. Their release kinetics have not been explored without confounding by heparin nor has their relationship to myocardial protein biomarkers. OBJECTIVE To compare ncRNA types in heparinase-treated samples with established and emerging protein biomarkers for myocardial injury. METHODS AND RESULTS Screening of 158 circRNAs and 21 lncRNAs in human cardiac tissue identified 12 circRNAs and 11 lncRNAs as potential biomarkers with cardiac origin. Eleven miRNAs were included. At low spike-in concentrations of myocardial tissue, significantly higher regression coefficients were observed across ncRNA types compared with cardiac troponins and cMyBP-C (cardiac myosin-binding protein C). Heparinase treatment of serial plasma and serum samples of patients undergoing transcoronary ablation of septal hypertrophy removed spurious correlations between miRNAs in non-heparinase-treated samples. After transcoronary ablation of septal hypertrophy, muscle-enriched miRNAs (miR-1 and miR-133a) showed a steeper and earlier increase than cardiac-enriched miRNAs (miR-499 and miR-208b). Putative cardiac lncRNAs, including LIPCAR (long intergenic noncoding RNA predicting cardiac remodeling and survival), did not rise, refuting a predominant cardiac origin. Cardiac circRNAs remained largely undetectable. In a validation cohort of acute myocardial infarction, receiver operating characteristic curve analysis revealed noninferiority of cardiac-enriched miRNAs, but miRNAs failed to identify cases presenting with low troponin values. cMyBP-C was validated as a biomarker with highly sensitive properties, and the combination of muscle-enriched miRNAs with high-sensitive cardiac troponin T and cMyBP-C returned the highest area under the curve values. CONCLUSIONS In a comparative assessment of ncRNAs and protein biomarkers for myocardial injury, cMyBP-C showed properties as the most sensitive cardiac biomarker while miRNAs emerged as promising candidates to integrate ncRNAs with protein biomarkers. Sensitivity of current miRNA detection is inferior to cardiac proteins but a multibiomarker combination of muscle-enriched miRNAs with cMyBP-C and cardiac troponins could open a new path of integrating complementary characteristics of different biomarker types.
Collapse
Affiliation(s)
- Christian Schulte
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Temo Barwari
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| | - Abhishek Joshi
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
- Bart’s Heart Centre, St. Bartholomew's Hospital, West Smithfield, London (A.J.)
| | - Konstantinos Theofilatos
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| | - Anna Zampetaki
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| | - Javier Barallobre-Barreiro
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| | - Bhawana Singh
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| | - Nils A. Sörensen
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Johannes T. Neumann
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Centre Hamburg Eppendorf, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Germany (C.S., N.A.S., J.T.N., T.Z., D.W., S.B.)
| | - Michael Marber
- King’s British Heart Foundation Centre, King’s College London, Guy’s and St Thomas’ Hospitals, United Kingdom (M. Marber)
| | - Christoph Liebetrau
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany and German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany (C.L.)
| | - Manuel Mayr
- From the King’s British Heart Foundation Centre, King’s College London, United Kingdom (C.S., T.B., A.J., K.T., A.Z., J.B.-B., B.S., M. Mayr)
| |
Collapse
|
17
|
Chen XJ, Zhang W, Bian ZP, Wang ZM, Zhang J, Wu HF, Shao YF, Zhang JN, Zhao S. Cardiac Myosin-Binding Protein C Release Profile After Cardiac Surgery in Intensive Care Unit. Ann Thorac Surg 2019; 108:1195-1201. [PMID: 31034826 DOI: 10.1016/j.athoracsur.2019.03.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/10/2019] [Accepted: 03/25/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Cardiac surgical procedures produce iatrogenic myocardial cell injury with necrosis that result in an obligatory release of biomarkers. Cardiac myosin binding protein C (cMyBP-C) has recently emerged as a specific and sensitive biomarker in patients with acute myocardial injury. We therefore aimed to investigate the release profiles of cMyBP-C after cardiac surgical procedures. METHODS Enzyme-linked immunosorbent assay to detect blood cMyBP-C was established by using two monoclonal antibodies against N-terminus of human cMyBP-C. Consecutive patients undergoing cardiac operations (N = 151) were recruited in this study. Blood cMyBP-C was assayed preoperatively, at intensive care unit arrival (0 hour after the operation), at 2 to 48 hours, and before discharge. The characteristics and detailed surgical procedure were recorded. RESULTS The established immunoassay was capable of detecting human cMyBP-C (0 to 1000 ng/L). The released cMyBP-C peaked immediately after cardiac surgery (0 h), attaining 3.8-fold higher than before the operation, dropped abruptly within 24 hours, and stayed at a higher level until discharge. Postoperative cMyBP-C levels correlated positively with high-sensitivity cardiac troponin T (hs-cTnT), creatine kinase, myoglobin, and creatine kinase MB isoenzyme. Different cardiac surgical procedures were characterized by different levels of release of cardiac biomarkers. Isolated off-pump coronary artery bypass grafting was associated with the smaller amount of cMyBP-C release, whereas valve replacement/plasty surgery produced higher release, in particular the multiple-valve surgery. Both cMyBP-C and hs-cTnT correlated with surgical techniques, postoperative intensive care unit stay, and hospital stay. CONCLUSIONS Circulating cMyBP-C is a promising novel biomarker for evaluating cardiac surgical trauma in patients undergoing a cardiac operation.
Collapse
Affiliation(s)
- Xiang-Jian Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Ping Bian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ze-Mu Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Zhang
- The Translational Platform of Jiangsu Province Hospital, Nanjing Bottests Biotechnology Co, Ltd, Nanjing, China
| | - Heng-Fang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong-Feng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ji-Nan Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; The Translational Platform of Jiangsu Province Hospital, Nanjing Bottests Biotechnology Co, Ltd, Nanjing, China
| | - Sheng Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Kaier TE, Alaour B, Marber M. Cardiac Myosin-Binding Protein C-From Bench to Improved Diagnosis of Acute Myocardial Infarction. Cardiovasc Drugs Ther 2019; 33:221-230. [PMID: 30617437 PMCID: PMC6509074 DOI: 10.1007/s10557-018-6845-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chest pain is responsible for 6-10% of all presentations to acute healthcare providers. Triage is inherently difficult and heavily reliant on the quantification of cardiac Troponin (cTn), as a minority of patients with an ultimate diagnosis of acute myocardial infarction (AMI) present with clear diagnostic features such as ST-elevation on the electrocardiogram. Owing to slow release and disappearance of cTn, many patients require repeat blood testing or present with stable but elevated concentrations of the best available biomarker and are thus caught at the interplay of sensitivity and specificity.We identified cardiac myosin-binding protein C (cMyC) in coronary venous effluent and developed a high-sensitivity assay by producing an array of monoclonal antibodies and choosing an ideal pair based on affinity and epitope maps. Compared to high-sensitivity cardiac Troponin (hs-cTn), we demonstrated that cMyC appears earlier and rises faster following myocardial necrosis. In this review, we discuss discovery and structure of cMyC, as well as the migration from a comparably insensitive to a high-sensitivity assay facilitating first clinical studies. This assay was subsequently used to describe relative abundance of the protein, compare sensitivity to two high-sensitivity cTn assays and test diagnostic performance in over 1900 patients presenting with chest pain and suspected AMI. A standout feature was cMyC's ability to more effectively triage patients. This distinction is likely related to the documented greater abundance and more rapid release profile, which could significantly improve the early triage of patients with suspected AMI.
Collapse
Affiliation(s)
- Thomas E Kaier
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, 4th Floor Lambeth Wing, Westminster Bridge Road, London, SE1 7EH, UK.
| | - Bashir Alaour
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, 4th Floor Lambeth Wing, Westminster Bridge Road, London, SE1 7EH, UK
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, 4th Floor Lambeth Wing, Westminster Bridge Road, London, SE1 7EH, UK
| |
Collapse
|
19
|
Zamanian Azodi M, Rezaei Tavirani M, Rezaei Tavirani M. Compound-Protein Interaction Analysis in Condition Following Cardiac Arrest. Galen Med J 2018; 7:e1380. [PMID: 34466450 PMCID: PMC8344096 DOI: 10.22086/gmj.v0i0.1380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/16/2018] [Accepted: 11/22/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cardiac arrest (CA) and differentially expressed genes (DEGs) relative to postCA have attracted the attention of scientist to prevent damages, which threaten patients. In the present study, metabolites relevant to DEGs of post-CA condition investigated via protein-compound interaction to understand the pathological mechanisms in the human body. MATERIALS AND METHODS STITCH plug-in integrated into Cytoscape V.3.6.1 was used to detect the most significant interacting compounds relative to DEGs of pig's brain after 5 minutes' CA. The genes were obtained from the Gene Expression Omnibus database. The identified elements were considered for further evaluation and validation by literature survey. RESULT Findings indicate that biochemical compounds including magnesium, calcium, glucose, glycerol, hydrogen, chloride, sulfate, and estradiol interact with DEGs in the two up- and down-regulated networks. CONCLUSION The compounds interacting with DEGs are suitable subjects to analysis for re-regulation of the body after CA.
Collapse
Affiliation(s)
- Mona Zamanian Azodi
- Student Research Committee, Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
20
|
Marber M. The future of myocardial injury biomarkers in cardiovascular disease: looking beyond cardiac troponins. Cardiovasc Res 2018; 114:e39-e40. [PMID: 29897495 DOI: 10.1093/cvr/cvy071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Michael Marber
- The Rayne Institute, St Thomas' Hospital, 4th Floor Lambeth Wing, Westminster Bridge Road, London SE1 7EH, UK
| |
Collapse
|
21
|
iTRAQ analysis of a mouse acute myocardial infarction model reveals that vitamin D binding protein promotes cardiomyocyte apoptosis after hypoxia. Oncotarget 2017; 9:1969-1979. [PMID: 29416745 PMCID: PMC5788613 DOI: 10.18632/oncotarget.23025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022] Open
Abstract
The proteome profile changes after acute myocardial infarction (AMI) and the roles played by important protein species remain poorly understood. Here, we constructed a mouse AMI model by ligating the left coronary artery of male C57B/6J mice to investigate the molecular changes after AMI on the protein level. Total proteins of the left ventricle were extracted and quantitatively analyzed by isobaric tags using relative and absolute quantitation (iTRAQ) technologies. The transcript and protein levels of important genes were further validated using quantitative polymerase chain reaction and western blot. An oxygen and glucose deprivation/reperfusion cell model was constructed using H9C2 cells to further validate the expression patterns and functions of important proteins after hypoxia. Seven hundred seventy-six proteins were identified as differentially abundant proteins after AMI, of which 406 were accumulated, and 370 were reduced. Gene ontology enrichment analysis showed that the most enriched molecular function category terms were binding, including calcium ion biding, GTP binding, actin binding and lipid binding. The expression levels of vitamin D binding protein (VDBP) and its related proteins were increased in both left ventricular tissue and H9C2 cells after ischemia-hypoxia. Overexpression of VDBP in H9C2 cells reduced vitamin D receptor and promoted the cell apoptosis rate after hypoxia. Our data provided new insights into proteome profile changes after AMI and indicated that VDBP could promote cardiomyocyte apoptosis after hypoxia.
Collapse
|
22
|
Anand A, Chin C, Shah ASV, Kwiecinski J, Vesey A, Cowell J, Weber E, Kaier T, Newby DE, Dweck M, Marber MS, Mills NL. Cardiac myosin-binding protein C is a novel marker of myocardial injury and fibrosis in aortic stenosis. Heart 2017; 104:1101-1108. [PMID: 29196542 PMCID: PMC6031261 DOI: 10.1136/heartjnl-2017-312257] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 01/30/2023] Open
Abstract
Objective Cardiac myosin-binding protein C (cMyC) is an abundant sarcomeric protein and novel highly specific marker of myocardial injury. Myocyte death characterises the transition from hypertrophy to replacement myocardial fibrosis in advanced aortic stenosis. We hypothesised that serum cMyC concentrations would be associated with cardiac structure and outcomes in patients with aortic stenosis. Methods cMyC was measured in two cohorts in which serum had previously been prospectively collected: a mechanism cohort of patients with aortic stenosis (n=161) and healthy controls (n=46) who underwent cardiac MRI, and an outcome cohort with aortic stenosis (n=104) followed for a median of 11.3 years. Results In the mechanism cohort, cMyC concentration correlated with left ventricular mass (adjusted β=11.0 g/m2 per log unit increase in cMyC, P<0.001), fibrosis volume (adjusted β=8.0 g, P<0.001) and extracellular volume (adjusted β=1.3%, P=0.01) in patients with aortic stenosis but not in controls. In those with late gadolinium enhancement (LGE) indicative of myocardial fibrosis, cMyC concentrations were higher (32 (21–56) ng/L vs 17 (12–24) ng/L without LGE, P<0.001). cMyC was unrelated to coronary calcium scores. Unadjusted Cox proportional hazards analysis in the outcome cohort showed greater all-cause mortality (HR 1.49 per unit increase in log cMyC, 95% CI 1.11 to 2.01, P=0.009). Conclusions Serum cMyC concentration is associated with myocardial hypertrophy, fibrosis and an increased risk of mortality in aortic stenosis. The quantification of serum sarcomeric protein concentrations provides objective measures of disease severity and their clinical utility to monitor the progression of aortic stenosis merits further study. Clinical trial registration NCT1755936; Post-results.
Collapse
Affiliation(s)
- Atul Anand
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Calvin Chin
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Anoop S V Shah
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Jacek Kwiecinski
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.,First Department of Cardiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alex Vesey
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Joanna Cowell
- Department of Geriatric Medicine, Royal Victoria Building, Edinburgh, UK
| | - Ekkehard Weber
- Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Thomas Kaier
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marc Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Michael S Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Kaier TE, Twerenbold R, Puelacher C, Marjot J, Imambaccus N, Boeddinghaus J, Nestelberger T, Badertscher P, Sabti Z, Giménez MR, Wildi K, Hillinger P, Grimm K, Loeffel S, Shrestha S, Widmer DF, Cupa J, Kozhuharov N, Miró Ò, Martín-Sánchez FJ, Morawiec B, Rentsch K, Lohrmann J, Kloos W, Osswald S, Reichlin T, Weber E, Marber M, Mueller C. Direct Comparison of Cardiac Myosin-Binding Protein C With Cardiac Troponins for the Early Diagnosis of Acute Myocardial Infarction. Circulation 2017; 136:1495-1508. [PMID: 28972002 PMCID: PMC5642333 DOI: 10.1161/circulationaha.117.028084] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cardiac myosin-binding protein C (cMyC) is a cardiac-restricted protein that is more abundant than cardiac troponins (cTn) and is released more rapidly after acute myocardial infarction (AMI). We evaluated cMyC as an adjunct or alternative to cTn in the early diagnosis of AMI. METHODS Unselected patients (N=1954) presenting to the emergency department with symptoms suggestive of AMI, concentrations of cMyC, and high-sensitivity (hs) and standard-sensitivity cTn were measured at presentation. The final diagnosis of AMI was independently adjudicated using all available clinical and biochemical information without knowledge of cMyC. The prognostic end point was long-term mortality. RESULTS Final diagnosis was AMI in 340 patients (17%). Concentrations of cMyC at presentation were significantly higher in those with versus without AMI (median, 237 ng/L versus 13 ng/L, P<0.001). Discriminatory power for AMI, as quantified by the area under the receiver-operating characteristic curve (AUC), was comparable for cMyC (AUC, 0.924), hs-cTnT (AUC, 0.927), and hs-cTnI (AUC, 0.922) and superior to cTnI measured by a contemporary sensitivity assay (AUC, 0.909). The combination of cMyC with hs-cTnT or standard-sensitivity cTnI (but not hs-cTnI) led to an increase in AUC to 0.931 (P<0.0001) and 0.926 (P=0.003), respectively. Use of cMyC more accurately classified patients with a single blood test into rule-out or rule-in categories: Net Reclassification Improvement +0.149 versus hs-cTnT, +0.235 versus hs-cTnI (P<0.001). In early presenters (chest pain <3 h), the improvement in rule-in/rule-out classification with cMyC was larger compared with hs-cTnT (Net Reclassification Improvement +0.256) and hs-cTnI (Net Reclassification Improvement +0.308; both P<0.001). Comparing the C statistics, cMyC was superior to hs-cTnI and standard sensitivity cTnI (P<0.05 for both) and similar to hs-cTnT at predicting death at 3 years. CONCLUSIONS cMyC at presentation provides discriminatory power comparable to hs-cTnT and hs-cTnI in the diagnosis of AMI and may perform favorably in patients presenting early after symptom onset. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT00470587.
Collapse
Affiliation(s)
- Thomas E Kaier
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Raphael Twerenbold
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Christian Puelacher
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Jack Marjot
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Nazia Imambaccus
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Jasper Boeddinghaus
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Thomas Nestelberger
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Patrick Badertscher
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Zaid Sabti
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Maria Rubini Giménez
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Karin Wildi
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Petra Hillinger
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Karin Grimm
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Sarah Loeffel
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Samyut Shrestha
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Dayana Flores Widmer
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Janosch Cupa
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Nikola Kozhuharov
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Òscar Miró
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - F Javier Martín-Sánchez
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Beata Morawiec
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Katharina Rentsch
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Jens Lohrmann
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Wanda Kloos
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Stefan Osswald
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Tobias Reichlin
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Ekkehard Weber
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| | - Michael Marber
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.).
| | - Christian Mueller
- From King's College London BHF Centre, Rayne Institute, St Thomas' Hospital, London, UK (T.K., J.M., N.I., M.M.); Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland (R.T., C.P., J.B., T.N., P.B., Z.S., M.R.G., K.W., P.H., K.G., S.L., S.S., D.F.W., J.C., N.K., J.L., W.K., S.O., T.R., C.M.); Department of General and Interventional Cardiology, University Heart Center Hamburg, Germany (R.T., M.R.G.); Emergency Department, Centre for Biomedical Network Research on Rare Diseases Instituto de Salud Carlos III, Hospital del Mar-IMIM, Barcelona, Spain (K.W.); Emergency Department, Hospital Clinic, Barcelona, Spain (O.M.); Global Research in Acute Conditions Network (O.M., F.J.M.S., B.M., C.M.); Emergency Department, Hospital Clinico San Carlos, Madrid, Spain (F.J.M.S.); 2nd Cardiology Department, Zabrze, University Silesia, Katowice, Poland (B.M.); Laboratory Medicine, University Hospital Basel, Switzerland (K.R.); and Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Germany (E.W.)
| |
Collapse
|
24
|
Datta K, Basak T, Varshney S, Sengupta S, Sarkar S. Quantitative proteomic changes during post myocardial infarction remodeling reveals altered cardiac metabolism and Desmin aggregation in the infarct region. J Proteomics 2017; 152:283-299. [DOI: 10.1016/j.jprot.2016.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/11/2016] [Accepted: 11/23/2016] [Indexed: 12/14/2022]
|
25
|
|
26
|
Lipps C, Nguyen JH, Pyttel L, Lynch TL, Liebetrau C, Aleshcheva G, Voss S, Dörr O, Nef HM, Möllmann H, Hamm CW, Sadayappan S, Troidl C. N-terminal fragment of cardiac myosin binding protein-C triggers pro-inflammatory responses in vitro. J Mol Cell Cardiol 2016; 99:47-56. [PMID: 27616755 DOI: 10.1016/j.yjmcc.2016.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/02/2016] [Accepted: 09/03/2016] [Indexed: 01/25/2023]
Abstract
Myocardial infarction (MI) leads to loss and degradation of contractile cardiac tissue followed by sterile inflammation of the myocardium through activation and recruitment of innate and adaptive cells of the immune system. Recently, it was shown that cardiac myosin binding protein-C (cMyBP-C), a protein of the cardiac sarcomere, is degraded following MI, releasing a predominant N-terminal 40-kDa fragment (C0C1f) into myocardial tissue and the systemic circulation. We hypothesized that early release of C0C1f contributes to the initiation of inflammation and plays a key role in recruitment and activation of immune cells. Therefore, we investigated the role of C0C1f on macrophage/monocyte activation using both mouse bone marrow-derived macrophages and human monocytes. Here we demonstrate that C0C1f leads to macrophage/monocyte activation in vitro. Furthermore, C0C1f induces strong upregulation of pro-inflammatory cytokines (interleukin-6 (IL-6), tumor necrosis factor α (TNFα), and interleukin-1β (IL-1β)) in cultured murine macrophages and human monocytes, resulting in a pro-inflammatory phenotype. We identified the toll-like receptor 4 (TLR4), toll-like receptor 2 (TLR2), and Advanced Glycosylation End Product-Specific Receptor (RAGE) as potential receptors for C0C1f whose activation leads to mobilization of the NFκB signaling pathway, a central mediator of the pro-inflammatory signaling cascade. Thus, C0C1f appears to be a key player in the initiation of inflammatory processes and might also play an important role upon MI.
Collapse
Affiliation(s)
- Christoph Lipps
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany
| | - Jenine H Nguyen
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany
| | - Lukas Pyttel
- Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany
| | - Thomas L Lynch
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| | | | | | - Sandra Voss
- Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany
| | - Oliver Dörr
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany
| | - Holger M Nef
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany
| | - Helge Möllmann
- Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany
| | - Christian W Hamm
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany; Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Christian Troidl
- Department of Experimental Cardiology, Medical Clinics I, Justus Liebig University, 35392 Giessen, Germany; Kerckhoff Heart and Thorax Center, 61231 Bad Nauheim, Germany.
| |
Collapse
|
27
|
Rather RA, Malik VS, Trikha D, Bhat O, Dhawan V. Aqueous Terminalia arjuna extract modulates expression of key atherosclerosis-related proteins in a hypercholesterolemic rabbit: A proteomic-based study. Proteomics Clin Appl 2016; 10:750-759. [PMID: 26934842 DOI: 10.1002/prca.201500114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/15/2016] [Accepted: 02/29/2016] [Indexed: 07/07/2024]
Abstract
PURPOSE The present study evaluates the effect of an aqueous extract of Terminalia arjuna (aqTAE) on protein expression in aortic plaques of hypercholesterolemic rabbits using a proteomic approach. EXPERIMENTAL DESIGN Thirty male New Zealand rabbits (n = 6) were employed as Gp1 (stock diet); Gp2 (high-fat diet [HFD]); Gp3 (stock diet + aqTAE); Gp4 (HFD + aqTAE); and Gp5 (HFD + atorvastatin) and followed for 6 months. Protein lysates of aortic tissues were separated by 2DE and proteins were identified by MALDI-TOF/MS. RESULTS Serum lipids were found to be significantly increased by an HFD and reduced by aqTAE both at 3 and 6 months (Gp4 vs. Gp2; p < 0.05). Total 79 spots were differentially expressed, among which 60 individual proteins were identified, 31 grouped as atherosclerosis-related proteins and 29 classified as others. aqTAE significantly attenuated the protein expression of tumor necrosis factor α, cyclooxygenase-2, MMP-9, HSP60, ICAM-5, Endothelin-3, Vimentin, Protein S100-A9 besides others. Many of the observed proteins are known to be consistently associated with endothelial dysfunction, inflammation, plaque rupture, and immune imbalance. CONCLUSIONS AND CLINICAL RELEVANCE Strong hypolipidemic effects of aqTAE and attenuation of these signature atherogenic biomarkers using proteomics highlights the fact that aqTAE may be useful in the prevention and management of atherosclerosis.
Collapse
Affiliation(s)
- Riyaz Ahmad Rather
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Vivek Singh Malik
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Dimple Trikha
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Owais Bhat
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| |
Collapse
|
28
|
Liu CH, Kuo SW, Hsu LM, Huang SC, Wang CH, Tsai PR, Chen YS, Jou TS, Ko WJ. Peroxiredoxin 1 induces inflammatory cytokine response and predicts outcome of cardiogenic shock patients necessitating extracorporeal membrane oxygenation: an observational cohort study and translational approach. J Transl Med 2016; 14:114. [PMID: 27142532 PMCID: PMC4855332 DOI: 10.1186/s12967-016-0869-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/18/2016] [Indexed: 12/02/2022] Open
Abstract
Background Extracellular peroxiredoxin 1 (Prdx1) has been implicated to play a pivotal role in regulating inflammation; however, its function in tissue hypoxia-induced inflammation, such as severe cardiogenic shock patients, has not yet been defined. Thus, the objective of this study was to test the hypothesis that Prdx1 possesses prognostic value and instigates systemic inflammatory response syndrome in cardiogenic shock patients undergoing extracorporeal membrane oxygenation (ECMO) support. Methods We documented the early time course evolution of circulatory Prdx1, hypoxic marker carbonic anhydrase IX, inflammatory cytokines including IL-6, IL-8, IL-10, MCP-1, TNF-α, IL-1β, and danger signaling receptors (TLR4 and CD14) in a cohort of cardiogenic shock patients within 1 day after ECMO support. In vitro investigations employing cultured murine macrophage cell lines and human monocytes were applied to clarify the relationship between Prdx1 and inflammatory response. Results Prdx1 not only peaked earlier than all the other cytokines we studied during the initial course, but also predicted a worse outcome in patients who had higher initial Prdx1 plasma levels. The Prdx1 levels in patients positively correlated with hypoxic markers carbonic anhydrase IX and lactate, and inflammatory cytokines. In vitro study demonstrated that hypoxia/reoxygenation induced Prdx1 release from human monocytes and enhanced the responsiveness of the monocytes in Prdx1-induced cytokine secretions. Furthermore, functional inhibition by Prdx1 antibody implicated a crucial role of Prdx1 in hypoxia/reoxygenation-induced IL-6 secretion. Conclusions Prdx1 release during the early phase of ECMO support in cardiogenic shock patients is associated with the development of systemic inflammatory response syndrome and poor clinical outcomes. Thus, circulating Prdx1 provides not only prognostic information but may be a promising target against ischemia/reperfusion injury. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0869-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chia-Hsiung Liu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.,Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Shuenn-Wen Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Li-Ming Hsu
- Department of Traumatology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Shu-Chien Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Chih-Hsien Wang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Pi-Ru Tsai
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Yih-Sharng Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Tzuu-Shuh Jou
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC. .,College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
| | - Wen-Je Ko
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC. .,Department of Traumatology, National Taiwan University Hospital, Taipei, Taiwan, ROC. .,College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
29
|
Marjot J, Liebetrau C, Goodson RJ, Kaier T, Weber E, Heseltine P, Marber MS. The development and application of a high-sensitivity immunoassay for cardiac myosin-binding protein C. Transl Res 2016; 170:17-25.e5. [PMID: 26713894 PMCID: PMC4796026 DOI: 10.1016/j.trsl.2015.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/14/2015] [Accepted: 11/28/2015] [Indexed: 11/20/2022]
Abstract
Cardiac troponins (cTns) are released and cleared slowly after myocardial injury. Cardiac myosin-binding protein C (cMyC) is a similar cardiac-restricted protein that has more rapid release and clearance kinetics. Direct comparisons are hampered by the lack of an assay for cMyC that matches the sensitivity of the contemporary assays for cTnI and cTnT. Using a novel pair of monoclonal antibodies, we generated a sensitive assay for MyC on the Erenna platform (Singulex) and compared serum concentrations with those of cTnI (Abbott) and cTnT (Roche) in stable ambulatory cardiac patients without evidence of acute cardiac injury or significant coronary artery stenoses. The assay for cMyC had a lower limit of detection of 0.4 ng/L, a lower limit of quantification (LLoQ) of 1.2 ng/L (LLoQ at 20% coefficient of variation [CV]) and reasonable recovery (107.1 ± 3.7%; mean ± standard deviation), dilutional linearity (101.0 ± 7.7%), and intraseries precision (CV, 11 ± 3%) and interseries precision (CV, 13 ± 3%). In 360 stable patients, cMyC was quantifiable in 359 patients and compared with cTnT and cTnI measured using contemporary high-sensitivity assays. cMyC concentration (median, 12.2 ng/L; interquartile range [IQR], 7.9-21.2 ng/L) was linearly correlated with those for cTnT (median, <3.0 ng/L; IQR, <3.0-4.9 ng/L; R = 0.56, P < 0.01) and cTnI (median, 2.10 ng/L; IQR, 1.3-4.2 ng/L; R = 0.77, P < 0.01) and showed similar dependencies on age, renal function, and left ventricular function. We have developed a high-sensitivity assay for cMyC. Concentrations of cMyC in clinically stable patients are highly correlated with those of cTnT and cTnI. This high correlation may enable ratiometric comparisons between biomarkers to distinguish clinical instability.
Collapse
Key Words
- acs, acute coronary syndrome
- ami, acute myocardial infarction
- cmyc, cardiac myosin–binding protein c
- ctn, cardiac troponin
- cv, coefficient of variation
- de, detected event
- lob, limit of blank
- lod, lower limit of detection
- lloq, lower limit of quantification
- mp, magnetic microparticle
- nste-acs, non–st-elevation acute coronary syndrome
Collapse
Affiliation(s)
- Jack Marjot
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Christoph Liebetrau
- Kerckhoff Heart and Thorax Center, Department of Cardiology, Bad Nauheim, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
| | | | - Thomas Kaier
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK
| | - Ekkehard Weber
- Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | - Michael S Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, London, UK.
| |
Collapse
|
30
|
Abstract
Calcified aortic stenosis is one of the most common causes of heart failure in the elderly. Current guidelines recommend aortic valve replacement in patients with severe disease and evidence of decompensation based on either symptoms or impaired systolic ejection fraction. However, symptoms are often subjective whilst impaired ejection fraction is not a sensitive marker of ventricular decompensation. Interest has surrounded the use of cardiac biochemical markers as objective measures of left ventricular decompensation in aortic stenosis. We will first examine mechanisms of release of biochemical markers associated with myocardial wall stress (BNP/NT-proBNP), myocardial fibrosis (markers of collagen metabolism, galectin-3, soluble ST2) and myocyte death/myocardial ischemia (high-sensitivity cardiac troponins, heart-type fatty acid binding protein, myosin-binding protein C); and discuss future directions of these markers.
Collapse
Affiliation(s)
- Calvin W L Chin
- a Department of Cardiovascular Medicine , National Heart Center Singapore , Singapore .,b Duke-NUS Graduate Medical School , Singapore
| | - Andie H Djohan
- c Barts and the London School of Medicine & Dentistry, Queen Mary, University of London , London , UK , and
| | - Chim C Lang
- d Division of Cardiovascular and Diabetes Medicine , University of Dundee , Dundee , UK
| |
Collapse
|
31
|
Cabrera-Fuentes HA, Alba-Alba C, Aragones J, Bernhagen J, Boisvert WA, Bøtker HE, Cesarman-Maus G, Fleming I, Garcia-Dorado D, Lecour S, Liehn E, Marber MS, Marina N, Mayr M, Perez-Mendez O, Miura T, Ruiz-Meana M, Salinas-Estefanon EM, Ong SB, Schnittler HJ, Sanchez-Vega JT, Sumoza-Toledo A, Vogel CW, Yarullina D, Yellon DM, Preissner KT, Hausenloy DJ. Meeting report from the 2nd International Symposium on New Frontiers in Cardiovascular Research. Protecting the cardiovascular system from ischemia: between bench and bedside. Basic Res Cardiol 2016; 111:7. [PMID: 26667317 PMCID: PMC4679108 DOI: 10.1007/s00395-015-0527-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 12/17/2022]
Abstract
Recent advances in basic cardiovascular research as well as their translation into the clinical situation were the focus at the last "New Frontiers in Cardiovascular Research meeting". Major topics included the characterization of new targets and procedures in cardioprotection, deciphering new players and inflammatory mechanisms in ischemic heart disease as well as uncovering microRNAs and other biomarkers as versatile and possibly causal factors in cardiovascular pathogenesis. Although a number of pathological situations such as ischemia-reperfusion injury or atherosclerosis can be simulated and manipulated in diverse animal models, also to challenge new drugs for intervention, patient studies are the ultimate litmus test to obtain unequivocal information about the validity of biomedical concepts and their application in the clinics. Thus, the open and bidirectional exchange between bench and bedside is crucial to advance the field of ischemic heart disease with a particular emphasis of understanding long-lasting approaches in cardioprotection.
Collapse
Affiliation(s)
- Hector A Cabrera-Fuentes
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
| | - Corina Alba-Alba
- Institute of Genetics, Univeristy of the Sea. Puerto Escondido Campus, Oaxaca Oaxacan System of State Universities (SUNEO), Oaxaca, México
| | - Julian Aragones
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, Madrid, Spain
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - William A Boisvert
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, Aarhus N, Denmark
| | | | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt, Germany
| | | | - Sandrine Lecour
- Hatter Institute and MRC Inter-University Cape Heart Unit, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Elisa Liehn
- Institute for Molecular Cardiovascular Research, RWTH University Hospital Aachen, Aachen, Germany
| | - Michael S Marber
- Department of Cardiology, The Rayne Institute, St Thomas' Campus, King's College London, London, UK
| | - Nephtali Marina
- Department of Clinical Pharmacology, University College London, London, UK
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Oscar Perez-Mendez
- Department of Molecular Biology, National Institute of Cardiology, Mexico City, Mexico
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marisol Ruiz-Meana
- Valld'Hebron University Hospital and Research Institute, Barcelona, Spain
| | | | - Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Hans J Schnittler
- Institute of Anatomy and Vascular Biology, Westfalian-Wilhelms-University, Münster, Germany
| | - Jose T Sanchez-Vega
- Laboratory of Parasitology, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adriana Sumoza-Toledo
- Laboratorio Multidisciplinario de Ciencias Biomédicas, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana campus Veracruz, Veracruz, Mexico
| | - Carl-Wilhelm Vogel
- Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Dina Yarullina
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore.
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
- The Hatter Cardiovascular Institute, University College London, London, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
32
|
Maznyczka A, Kaier T, Marber M. Troponins and other biomarkers in the early diagnosis of acute myocardial infarction. Postgrad Med J 2015; 91:322-30. [DOI: 10.1136/postgradmedj-2014-133129] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 05/08/2015] [Indexed: 12/24/2022]
|
33
|
Basak T, Varshney S, Akhtar S, Sengupta S. Understanding different facets of cardiovascular diseases based on model systems to human studies: a proteomic and metabolomic perspective. J Proteomics 2015; 127:50-60. [PMID: 25956427 DOI: 10.1016/j.jprot.2015.04.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/08/2015] [Accepted: 04/25/2015] [Indexed: 02/02/2023]
Abstract
UNLABELLED Cardiovascular disease has remained as the largest cause of morbidity and mortality worldwide. From dissecting the disease aetiology to identifying prognostic markers for better management of the disease is still a challenge for researchers. In the post human genome sequencing era much of the thrust has been focussed towards application of advanced genomic tools along with evaluation of traditional risk factors. With the advancement of next generation proteomics and metabolomics approaches it has now become possible to understand the protein interaction network & metabolic rewiring which lead to the perturbations of the disease phenotype. Further, elucidating different post translational modifications using advanced mass spectrometry based methods have provided an impetus towards in depth understanding of the proteome. The past decade has observed a plethora of studies where proteomics has been applied successfully to identify potential prognostic and diagnostic markers as well as to understand the disease mechanisms for various types of cardiovascular diseases. In this review, we attempted to document relevant proteomics based studies that have been undertaken either to identify potential biomarkers or have elucidated newer mechanistic insights into understanding the patho-physiology of cardiovascular disease, primarily coronary artery disease, cardiomyopathy, and myocardial ischemia. We have also provided a perspective on the potential of proteomics in combating this deadly disease. BIOLOGICAL SIGNIFICANCE This review has catalogued recent studies on proteomics and metabolomics involved in understanding several cardiovascular diseases (CVDs). A holistic systems biology based approach, of which proteomics and metabolomics are two very important components, would help in delineating various pathways associated with complex disorders like CVD. This would ultimately provide better mechanistic understanding of the disease biology leading to development of prognostic biomarkers. This article is part of a Special Issue entitled: Proteomics in India.
Collapse
Affiliation(s)
- Trayambak Basak
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India.
| | - Swati Varshney
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India
| | - Shamima Akhtar
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India
| | - Shantanu Sengupta
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110020, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IGIB South Campus, New Delhi, India.
| |
Collapse
|
34
|
Smith LE, White MY. The role of post-translational modifications in acute and chronic cardiovascular disease. Proteomics Clin Appl 2015; 8:506-21. [PMID: 24961403 DOI: 10.1002/prca.201400052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 05/27/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) in one of the leading causes of mortality and morbidity worldwide, accounting for both primary diseases of the heart and vasculature and arising as a co-morbidity with numerous pathologies, including type 2 diabetes mellitus (T2DM). There has been significant emphasis on the role of the genome in CVD, aiding in the definition of 'at-risk' patients. The extent of disease penetrance however, can be influenced by environmental factors that are not detectable by investigating the genome alone. By targeting the transcriptome in response to CVD, the interplay between genome and environment is more apparent, however this implies the level of protein expression without reference to proteolytic turnover, or potentially more importantly, without defining the role of PTMs in the development of disease. Here, we discuss the role of both brief and irreversible PTMs in the setting of myocardial ischemia/reperfusion injury. Key proteins involved in calcium regulation have been observed as differentially modified by phosphorylation/O-GlcNAcylation or phosphorylation/redox modifications, with the level of interplay dependent on the physiological or pathophysiological state. The ability to modify crucial sites to produce the desired functional output is modulated by the presence of other PTMs as exemplified in the T2DM heart, where hyperglycemia results in aberrant O-GlcNAcylation and advanced glycation end products. By using the signalling events predicted to be critical to post-conditioning, an intervention with great promise for the cardioprotection of the ischemia/reperfusion injured heart, as an example, we discuss the level of PTMs and their interplay. The inability of post-conditioning to protect the diabetic heart may be regulated by aberrant PTMs influencing those sites necessary for protection.
Collapse
Affiliation(s)
- Lauren E Smith
- Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
35
|
Baker JO, Tyther R, Liebetrau C, Clark J, Howarth R, Patterson T, Möllmann H, Nef H, Sicard P, Kailey B, Devaraj R, Redwood SR, Kunst G, Weber E, Marber MS. Cardiac myosin-binding protein C: a potential early biomarker of myocardial injury. Basic Res Cardiol 2015; 110:23. [PMID: 25837837 PMCID: PMC4383815 DOI: 10.1007/s00395-015-0478-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/11/2015] [Accepted: 03/11/2015] [Indexed: 11/28/2022]
Abstract
Cardiac troponins are released and cleared slowly after myocardial injury, complicating the diagnosis of early, and recurrent, acute myocardial infarction. Cardiac myosin-binding protein C (cMyC) is a similarly cardiac-restricted protein that may have different release/clearance kinetics. Using novel antibodies raised against the cardiac-specific N-terminus of cMyC, we used confocal microscopy, immunoblotting and immunoassay to document its location and release. In rodents, we demonstrate rapid release of cMyC using in vitro and in vivo models of acute myocardial infarction. In patients, with ST elevation myocardial infarction (STEMI, n = 20), undergoing therapeutic ablation of septal hypertrophy (TASH, n = 20) or having coronary artery bypass surgery (CABG, n = 20), serum was collected prospectively and frequently. cMyC appears in the serum as full-length and fragmented protein. Compared to cTnT measured using a contemporary high-sensitivity commercial assay, cMyC peaks earlier (STEMI, 9.3 ± 3.1 vs 11.8 ± 3.4 h, P < 0.007; TASH, 9.7 ± 1.4 vs 21.6 ± 1.4 h, P < 0.0001), accumulates more rapidly (during first 4 h after TASH, 25.8 ± 1.9 vs 4.0 ± 0.4 ng/L/min, P < 0.0001) and disappears more rapidly (post-CABG, decay half-time 5.5 ± 0.8 vs 22 ± 5 h, P < 0.0001). Our results demonstrate that following defined myocardial injury, the rise and fall in the serum of cMyC is more rapid than that of cTnT. We speculate that these characteristics could enable earlier diagnosis of myocardial infarction and reinfarction in suspected non-STEMI, a population not included in this early translational study.
Collapse
Affiliation(s)
- James O Baker
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, 4th Floor Lambeth Wing, Westminster Bridge Road, London, SE1 7EH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mitra A, Basak T, Ahmad S, Datta K, Datta R, Sengupta S, Sarkar S. Comparative Proteome Profiling during Cardiac Hypertrophy and Myocardial Infarction Reveals Altered Glucose Oxidation by Differential Activation of Pyruvate Dehydrogenase E1 Component Subunit β. J Mol Biol 2014; 427:2104-20. [PMID: 25451023 DOI: 10.1016/j.jmb.2014.10.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/09/2014] [Accepted: 10/29/2014] [Indexed: 12/19/2022]
Abstract
Cardiac hypertrophy and myocardial infarction (MI) are two etiologically different disease forms with varied pathological characteristics. However, the precise molecular mechanisms and specific causal proteins associated with these diseases are obscure to date. In this study, a comparative cardiac proteome profiling was performed in Wistar rat models for diseased and control (sham) groups using two-dimensional difference gel electrophoresis followed by matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry. Proteins were identified using Protein Pilot™ software (version 4.0) and were subjected to stringent statistical analysis. Alteration of key proteins was validated by Western blot analysis. The differentially expressed protein sets identified in this study were associated with different functional groups, involving various metabolic pathways, stress responses, cytoskeletal organization, apoptotic signaling and other miscellaneous functions. It was further deciphered that altered energy metabolism during hypertrophy in comparison to MI may be predominantly attributed to induced glucose oxidation level, via reduced phosphorylation of pyruvate dehydrogenase E1 component subunit β (PDHE1-B) protein during hypertrophy. This study reports for the first time the global changes in rat cardiac proteome during two etiologically different cardiac diseases and identifies key signaling regulators modulating ontogeny of these two diseases culminating in heart failure. This study also pointed toward differential activation of PDHE1-B that accounts for upregulation of glucose oxidation during hypertrophy. Downstream analysis of altered proteome and the associated modulators would enhance our present knowledge regarding altered pathophysiology of these two etiologically different cardiac disease forms.
Collapse
Affiliation(s)
- Arkadeep Mitra
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Trayambak Basak
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Shadab Ahmad
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Kaberi Datta
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Ritwik Datta
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Shantanu Sengupta
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Sagartirtha Sarkar
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India.
| |
Collapse
|
37
|
Lynch TL, Sadayappan S. Surviving the infarct: A profile of cardiac myosin binding protein-C pathogenicity, diagnostic utility, and proteomics in the ischemic myocardium. Proteomics Clin Appl 2014; 8:569-77. [PMID: 24888514 PMCID: PMC4162529 DOI: 10.1002/prca.201400011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/31/2014] [Accepted: 05/20/2014] [Indexed: 12/17/2022]
Abstract
Cardiac myosin binding protein-C (cMyBP-C) is a regulatory protein of the contractile apparatus within the cardiac sarcomere. Ischemic injury to the heart during myocardial infarction (MI) results in the cleavage of cMyBP-C in a phosphorylation-dependent manner and release of an N-terminal fragment (C0C1f) into the circulation. C0C1f has been shown to be pathogenic within cardiac tissue, leading to the development of heart failure. Based on its high levels and early release into the circulation post-MI, C0C1f may serve as a novel biomarker for diagnosing MI more effectively than current clinically used biomarkers. Over time, circulating C0C1f could trigger an autoimmune response leading to myocarditis and progressive cardiac dysfunction. Given the importance of cMyBP-C phosphorylation state in the context of proteolytic cleavage and release into the circulation post-MI, understanding the posttranslational modifications (PTMs) of cMyBP-C would help in further elucidating the role of this protein in health and disease. Accordingly, recent studies have implemented the latest proteomics approaches to define the PTMs of cMyBP-C. The use of such proteomics assays may provide accurate quantitation of the levels of cMyBP-C in the circulation following MI, which could, in turn, demonstrate the efficacy of using plasma cMyBP-C as a cardiac-specific early biomarker of MI. In this review, we define the pathogenic and potential immunogenic effects of C0C1f on cardiac function in the post-MI heart. We also discuss the most advanced proteomics approaches now used to determine cMyBP-C PTMs with the aim of validating C0C1f as an early biomarker of MI.
Collapse
Affiliation(s)
- Thomas L Lynch
- Division of Cardiology, Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | | |
Collapse
|
38
|
Cuello F, Shankar-Hari M, Mayr U, Yin X, Marshall M, Suna G, Willeit P, Langley SR, Jayawardhana T, Zeller T, Terblanche M, Shah AM, Mayr M. Redox state of pentraxin 3 as a novel biomarker for resolution of inflammation and survival in sepsis. Mol Cell Proteomics 2014; 13:2545-57. [PMID: 24958171 PMCID: PMC4188985 DOI: 10.1074/mcp.m114.039446] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In an endotoxaemic mouse model of sepsis, a tissue-based proteomics approach for biomarker discovery identified long pentraxin 3 (PTX3) as the lead candidate for inflamed myocardium. When the redox-sensitive oligomerization state of PTX3 was further investigated, PTX3 accumulated as an octamer as a result of disulfide-bond formation in heart, kidney, and lung—common organ dysfunctions seen in patients with sepsis. Oligomeric moieties of PTX3 were also detectable in circulation. The oligomerization state of PTX3 was quantified over the first 11 days in critically ill adult patients with sepsis. On admission day, there was no difference in the oligomerization state of PTX3 between survivors and non-survivors. From day 2 onward, the conversion of octameric to monomeric PTX3 was consistently associated with a greater survival after 28 days of follow-up. For example, by day 2 post-admission, octameric PTX3 was barely detectable in survivors, but it still constituted more than half of the total PTX3 in non-survivors (p < 0.001). Monomeric PTX3 was inversely associated with cardiac damage markers NT-proBNP and high-sensitivity troponin I and T. Relative to the conventional measurements of total PTX3 or NT-proBNP, the oligomerization of PTX3 was a superior predictor of disease outcome.
Collapse
Affiliation(s)
- Friederike Cuello
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK; §Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Centre, University Medical Center Hamburg-Eppendorf, Hamburg, 20246 Germany; ¶DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Manu Shankar-Hari
- ‖Critical Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH UK; **Division of Asthma Allergy and Lung Biology, King's College, London SE1 9RT, UK
| | - Ursula Mayr
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Xiaoke Yin
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Melanie Marshall
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Gonca Suna
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Peter Willeit
- ‡‡Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK; §§Department of Neurology, Innsbruck Medical University, Innsbruck, 6020 Austria
| | - Sarah R Langley
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Tamani Jayawardhana
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Tanja Zeller
- ¶¶Clinic for General and Interventional Cardiology, University Heart Centre Hamburg, Hamburg 20246, Germany
| | - Marius Terblanche
- ‖Critical Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH UK
| | - Ajay M Shah
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK
| | - Manuel Mayr
- From the ‡King's British Heart Foundation Centre, King's College London, SE5 9NU London, UK;
| |
Collapse
|
39
|
Kristensen LP, Larsen MR, Mickley H, Saaby L, Diederichsen AC, Lambrechtsen J, Rasmussen LM, Overgaard M. Plasma proteome profiling of atherosclerotic disease manifestations reveals elevated levels of the cytoskeletal protein vinculin. J Proteomics 2014; 101:141-53. [DOI: 10.1016/j.jprot.2013.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 11/16/2022]
|
40
|
Marshall KD, Edwards MA, Krenz M, Davis JW, Baines CP. Proteomic mapping of proteins released during necrosis and apoptosis from cultured neonatal cardiac myocytes. Am J Physiol Cell Physiol 2014; 306:C639-47. [PMID: 24401845 DOI: 10.1152/ajpcell.00167.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cardiac injury induces myocyte apoptosis and necrosis, resulting in the secretion and/or release of intracellular proteins. Currently, myocardial injury can be detected by analysis of a limited number of biomarkers in blood or coronary artery perfusate. However, the complete proteomic signature of protein release from necrotic cardiac myocytes is unknown. Therefore, we undertook a proteomic-based study of proteins released from cultured neonatal rat cardiac myocytes in response to H2O2 (necrosis) or staurosporine (apoptosis) to identify novel specific markers of cardiac myocyte cell death. Necrosis and apoptosis resulted in the identification of 147 and 79 proteins, respectively. Necrosis resulted in a relative increase in the amount of many proteins including the classical necrotic markers lactate dehydrogenase (LDH), high-mobility group B1 (HMGB1), myoglobin, enolase, and 14-3-3 proteins. Additionally, we identified several novel markers of necrosis including HSP90, α-actinin, and Trim72, many of which were elevated over control levels earlier than classical markers of necrotic injury. In contrast, the majority of identified proteins remained at low levels during apoptotic cell death, resulting in no candidate markers for apoptosis being identified. Blotting for a selection of these proteins confirmed their release during necrosis but not apoptosis. We were able to confirm the presence of classical necrotic markers in the extracellular milieu of necrotic myocytes. We also were able to identify novel markers of necrotic cell death with relatively early release profiles compared with classical protein markers of necrosis. These results have implications for the discovery of novel biomarkers of necrotic myocyte injury, especially in the context of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Kurt D Marshall
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | | | | | | | | |
Collapse
|
41
|
Kakimoto Y, Ito S, Abiru H, Kotani H, Ozeki M, Tamaki K, Tsuruyama T. Sorbin and SH3 domain-containing protein 2 is released from infarcted heart in the very early phase: proteomic analysis of cardiac tissues from patients. J Am Heart Assoc 2013; 2:e000565. [PMID: 24342996 PMCID: PMC3886759 DOI: 10.1161/jaha.113.000565] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Few proteomic studies have examined human cardiac tissue following acute lethal infarction. Here, we applied a novel proteomic approach to formalin-fixed, paraffin-embedded human tissue and aimed to reveal the molecular changes in the very early phase of acute myocardial infarction. METHODS AND RESULTS Heart tissue samples were collected from 5 patients who died within 7 hours of myocardial infarction and from 5 age- and sex-matched control cases. Infarcted and control myocardia were histopathologically diagnosed and captured using laser microdissection. Proteins were extracted using an originally established method and analyzed using liquid chromatography-tandem mass spectrometry. The label-free quantification demonstrated that the levels of 21 proteins differed significantly between patients and controls. In addition to known biomarkers, the sarcoplasmic protein sorbin and SH3 domain-containing protein 2 (SORBS2) was greatly reduced in infarcted myocardia. Immunohistochemical analysis of cardiac tissues confirmed the decrease, and Western blot analysis showed a significant increase in serum sorbin and SH3 domain-containing protein 2 in acute myocardial infarction patients (n=10) compared with control cases (n=11). CONCLUSIONS Our advanced comprehensive analysis using patient tissues and serums indicated that sarcoplasmic sorbin and SH3 domain-containing protein 2 is released from damaged cardiac tissue into the bloodstream upon lethal acute myocardial infarction. The proteomic strategy presented here is based on precise microscopic findings and is quite useful for candidate biomarker discovery using human tissue samples stored in depositories.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Patel BG, Wilder T, Solaro RJ. Novel control of cardiac myofilament response to calcium by S-glutathionylation at specific sites of myosin binding protein C. Front Physiol 2013; 4:336. [PMID: 24312057 PMCID: PMC3834529 DOI: 10.3389/fphys.2013.00336] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/01/2013] [Indexed: 12/31/2022] Open
Abstract
Our previous studies demonstrated a relation between glutathionylation of cardiac myosin binding protein C (cMyBP-C) and diastolic dysfunction in a hypertensive mouse model stressed by treatment with salt, deoxycorticosterone acetate, and unilateral nephrectomy. Although these results strongly indicated an important role for S-glutathionylation of myosin binding protein C as a modifier of myofilament function, indirect effects of other post-translational modifications may have occurred. Moreover, we did not determine the sites of thiol modification by glutathionylation. To address these issues, we developed an in vitro method to mimic the in situ S-glutathionylation of myofilament proteins and determined direct functional effects and sites of oxidative modification employing Western blotting and mass spectrometry. We induced glutathionylation in vitro by treatment of isolated myofibrils and detergent extracted fiber bundles (skinned fibers) with oxidized glutathione (GSSG). Immuno-blotting results revealed increased glutathionylation with GSSG treatment of a protein band around 140 kDa. Using tandem mass spectrometry, we identified the 140 kDa band as cMyBP-C and determined the sites of glutathionylation to be at cysteines 655, 479, and 627. Determination of the relation between Ca2+-activation of myofibrillar acto-myosin ATPase rate demonstrated an increased Ca2+-sensitivity induced by the S-glutathionylation. Force generating skinned fiber bundles also showed an increase in Ca-sensitivity when treated with oxidized glutathione, which was reversed with the reducing agent, dithiothreitol (DTT). Our data demonstrate that a specific and direct effect of S-glutathionylation of myosin binding protein C is a significant increase in myofilament Ca2+-sensitivity. Our data also provide new insights into the functional significance of oxidative modification of myosin binding protein C and the potential role of domains not previously considered to be functionally significant as controllers of myofilament Ca2+-responsiveness and dynamics.
Collapse
Affiliation(s)
- Bindiya G Patel
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago Chicago, IL USA
| | | | | |
Collapse
|
43
|
Govindan S, Kuster DWD, Lin B, Kahn DJ, Jeske WP, Walenga JM, Leya F, Hoppensteadt D, Fareed J, Sadayappan S. Increase in cardiac myosin binding protein-C plasma levels is a sensitive and cardiac-specific biomarker of myocardial infarction. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2013; 3:60-70. [PMID: 23785583 PMCID: PMC3683403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/20/2013] [Indexed: 06/02/2023]
Abstract
Earlier studies have shown that cardiac myosin binding protein-C (cMyBP-C) is easily releasable into the circulation following myocardial infarction (MI) in animal models and patients. However, since its release kinetics has not been clearly demonstrated, no parameters are available to judge its efficacy as a bona fide biomarker of MI in patients with MI. To make this assessment, plasma levels of cMyBP-C and six known biomarkers of MI were determined by sandwich enzyme-linked immunosorbent assay in patients with MI who had before and after Percutaneous Transcoronary Angioplasty (PTCA), as well as healthy controls. Compared to healthy controls (22.3 ± 2.4 ng/mL (n=54)), plasma levels of cMyBP-C were significantly increased in patients with MI (105.1 ± 8.8 ng/mL (n=65), P<0.001). Out of 65 patients, 24 had very high levels of plasma cMyBP-C (116.5 ± 13.3 ng/mL), indicating high probability of MI. Importantly, cMyBP-C levels were significantly decreased in patients (n=40) at 12 hours post-PTCA (41.2 ± 9.3 ng/mL, P<0.001), compared to the patients with MI. Receiver operating characteristic analysis revealed that a plasma cMyBP-C reading of 68.1 ng/mL provided a sensitivity of 66.2% and a specificity of 100%. Also, myoglobin, carbonic anhydrase and creatine kinase-MB levels were significantly increased in MI patients who also had higher cMyBP-C levels. In contrast, levels of cardiac troponin I, glycogen phosphorylase and heart-type fatty acid binding protein were not significantly changed in the samples, indicating the importance of evaluating the differences in release kinetics of these biomarkers in the context of accurate diagnosis. Our findings suggest that circulating cMyBP-C is a sensitive and cardiac-specific biomarker with potential utility for the accurate diagnosis of MI.
Collapse
Affiliation(s)
- Suresh Govindan
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Diederik WD Kuster
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Brian Lin
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Daniel J Kahn
- Departments of Pathology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Pharmacology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Walter P Jeske
- Departments of Pathology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Thoracic and Cardiovascular Surgery, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Pharmacology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Jeanine M Walenga
- Departments of Pathology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Thoracic and Cardiovascular Surgery, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Pharmacology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Fred Leya
- Interventional Cardiology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Debra Hoppensteadt
- Departments of Pathology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Pharmacology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Jawed Fareed
- Departments of Pathology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
- Pharmacology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University ChicagoMaywood, IL 60153, USA
| |
Collapse
|
44
|
Eberini I, Wait R, Calabresi L, Sensi C, Miller I, Gianazza E. A proteomic portrait of atherosclerosis. J Proteomics 2013; 82:92-112. [DOI: 10.1016/j.jprot.2013.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 01/11/2023]
|
45
|
Mayr M, Zampetaki A, Willeit P, Willeit J, Kiechl S. MicroRNAs within the continuum of postgenomics biomarker discovery. Arterioscler Thromb Vasc Biol 2013; 33:206-14. [PMID: 23325478 DOI: 10.1161/atvbaha.112.300141] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/21/2012] [Indexed: 12/14/2022]
Abstract
The postgenomic shift in paradigm from reductionism to systems-wide network inference has increased recognition that cardiovascular diseases are not simply determined by the genome but arise from an interaction and dynamic dysregulation of gene regulatory networks, proteins, and metabolic alterations. The advent of postgenomic technologies promises to interrogate these complex pathophysiological perturbations by applying concepts of systemic relationships to biomarker discovery. A multibiomarker panel consisting of biomarkers capturing different levels of information (eg, microRNAs to assess endothelial and platelet activation, molecular lipid species to profile metabolic status, and proteolytic degradation products to assess vascular integrity) could outperform inflammatory biomarkers without vascular specificity in their ability of predicting cardiovascular risk. As atherosclerosis develops over decades, different biomarkers may be required for different stages of disease. Thus far, there is no simple blood test to directly assess the health of blood vessels or identify vulnerable patients. We discuss strategies for biomarker discovery using post genomics technologies, with a particular focus on circulating microRNAs. The aim is to reveal distinctive cardiovascular phenotypes and identify biomarker signatures that complement the Framingham risk scores in clinical decision-making and in a stratified medicine approach for early preventive treatment of disease.
Collapse
Affiliation(s)
- Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom.
| | | | | | | | | |
Collapse
|
46
|
Dong SY, Sun XN, Zeng Q, Xu Y, Sun J, Ma LH. Proteomic analysis of adverse outcomes in patients with acute coronary syndromes. Clin Chim Acta 2013. [DOI: 10.1016/j.cca.2012.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
47
|
Protein target quantification decision tree. INTERNATIONAL JOURNAL OF PROTEOMICS 2013; 2013:701247. [PMID: 23401774 PMCID: PMC3562589 DOI: 10.1155/2013/701247] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/10/2012] [Accepted: 12/18/2012] [Indexed: 11/17/2022]
Abstract
The utility of mass spectrometry-(MS-) based proteomic platforms and their clinical applications have become an emerging field in proteomics in recent years. Owing to its selectivity and sensitivity, MS has become a key technological platform in proteomic research. Using this platform, a large number of potential biomarker candidates for specific diseases have been reported. However, due to lack of validation, none has been approved for use in clinical settings by the Food and Drug Administration (FDA). Successful candidate verification and validation will facilitate the development of potential biomarkers, leading to better strategies for disease diagnostics, prognostics, and treatment. With the recent new developments in mass spectrometers, high sensitivity, high resolution, and high mass accuracy can be achieved. This greatly enhances the capabilities of protein biomarker validation. In this paper, we describe and discuss recent developments and applications of targeted proteomics methods for biomarker validation.
Collapse
|
48
|
Recent advances in cardiovascular proteomics. J Proteomics 2012; 81:3-14. [PMID: 23153792 DOI: 10.1016/j.jprot.2012.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 10/10/2012] [Accepted: 10/31/2012] [Indexed: 01/08/2023]
Abstract
Cardiovascular diseases (CVDs) are the major source of global morbidity and death and more people die annually from CVDs than from any other cause. These diseases can occur quickly, as seen in acute myocardial infarction (AMI), or progress slowly over years as with chronic heart failure. Advances in mass spectrometry detection and analysis, together with improved isolation and enrichment techniques allowing for the separation of organelles and membrane proteins, now allow for the indepth analysis of the cardiac proteome. Here we outline current insights that have been provided through cardiovascular proteomics, and discuss studies that have developed innovative technologies which permit the examination of the protein complement in specific organelles including exosomes and secreted proteins. We highlight these foundational studies and illustrate how they are providing the technologies and tools which are now being applied to further study cardiovascular disease; provide new diagnostic markers and potentially new methods of cardiac patient management with identification of novel drug targets. This article is part of a Special Issue entitled: From protein structures to clinical applications.
Collapse
|
49
|
Chugh S, Sharma P, Kislinger T, Gramolini AO. Clinical proteomics: getting to the heart of the matter. ACTA ACUST UNITED AC 2012; 5:377. [PMID: 22715282 DOI: 10.1161/circgenetics.110.957761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Shaan Chugh
- Department of Physiology, University of Toronto, ON, Canada
| | | | | | | |
Collapse
|
50
|
Marston SB, Gautel M. Introducing a series of topical special issues of the Journal of Muscle Research and Cell Motility: MYBPC3 special issue editorial. J Muscle Res Cell Motil 2012; 33:1-3. [PMID: 22555918 DOI: 10.1007/s10974-012-9296-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|